1
|
Serrano MV, Cottier S, Wang L, Moreira-Antepara S, Nzessi A, Liu Z, Williams B, Lee M, Schneiter R, Liu J. The C. elegans LON-1 protein requires its CAP domain for function in regulating body size and BMP signaling. Genetics 2025; 229:iyae202. [PMID: 39657262 PMCID: PMC11796460 DOI: 10.1093/genetics/iyae202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/19/2024] [Accepted: 11/25/2024] [Indexed: 12/17/2024] Open
Abstract
The CAP (cysteine-rich secretory proteins, antigen-5, and pathogenesis-related) proteins are widely expressed and have been implicated to play diverse roles ranging from mammalian reproduction to plant immune response. Increasing evidence supports a role of CAP proteins in lipid binding. The Caenorhabditis elegans CAP protein LON-1 is known to regulate body size and bone morphogenetic protein (BMP) signaling. LON-1 is a secreted protein with a conserved CAP domain and a C-terminal unstructured domain with no homology to other proteins. In this study, we report that the C-terminal domain of LON-1 is dispensable for its function. Instead, key conserved residues located in the CAP domain are critical for LON-1 function in vivo. We further showed that LON-1 is capable of binding sterol, but not fatty acid, in vitro, and that certain key residues implicated in LON-1 function in vivo are also important for LON-1 sterol binding in vitro. These findings suggest a role of LON-1 in regulating body size and BMP signaling via sterol binding.
Collapse
Affiliation(s)
| | - Stéphanie Cottier
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Lianzijun Wang
- Department of Biology, Baylor University, Waco, TX 76798, USA
| | | | - Anthony Nzessi
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Zhiyu Liu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Byron Williams
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Myeongwoo Lee
- Department of Biology, Baylor University, Waco, TX 76798, USA
| | - Roger Schneiter
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Jun Liu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
2
|
Zhebrun A, Ni JZ, Corveleyn L, Ghosh Roy S, Sidoli S, Gu SG. Two H3K23 histone methyltransferases, SET-32 and SET-21, function synergistically to promote nuclear RNAi-mediated transgenerational epigenetic inheritance in Caenorhabditis elegans. Genetics 2025; 229:iyae206. [PMID: 39661453 PMCID: PMC11796467 DOI: 10.1093/genetics/iyae206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024] Open
Abstract
Nuclear RNAi in Caenorhabditis elegans induces a set of transgenerationally heritable marks of H3K9me3, H3K23me3, and H3K27me3 at the target genes. The function of H3K23me3 in the nuclear RNAi pathway is largely unknown due to the limited knowledge of H3K23 histone methyltransferase (HMT). In this study we identified SET-21 as a novel H3K23 HMT. By taking combined genetic, biochemical, imaging, and genomic approaches, we found that SET-21 functions synergistically with a previously reported H3K23 HMT SET-32 to deposit H3K23me3 at the native targets of germline nuclear RNAi. We identified a subset of native nuclear RNAi targets that are transcriptionally activated in the set-21;set-32 double mutant. SET-21 and SET-32 are also required for robust transgenerational gene silencing induced by exogenous dsRNA. The set-21;set-32 double mutant strain exhibits an enhanced temperature-sensitive mortal germline phenotype compared to the set-32 single mutant, while the set-21 single mutant animals are fertile. We also found that HRDE-1 and SET-32 are required for cosuppression, a transgene-induced gene silencing phenomenon, in C. elegans germline. Together, these results support a model in which H3K23 HMTs SET-21 and SET-32 function cooperatively as germline nuclear RNAi factors and promote the germline immortality under the heat stress.
Collapse
Affiliation(s)
- Anna Zhebrun
- Department of Molecular Biology and Biochemistry, Rutgers The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Julie Z Ni
- Department of Molecular Biology and Biochemistry, Rutgers The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Laura Corveleyn
- Laboratory of Pharmaceutical Biotechnology, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, Gent 9000, Belgium
| | - Siddharth Ghosh Roy
- Department of Molecular Biology and Biochemistry, Rutgers The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Simone Sidoli
- Department of Chemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Sam G Gu
- Department of Molecular Biology and Biochemistry, Rutgers The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA
| |
Collapse
|
3
|
Shugarts Devanapally NM, Sathya A, Yi AL, Chan WM, Marre JA, Jose AM. Intergenerational transport of double-stranded RNA in C. elegans can limit heritable epigenetic changes. eLife 2025; 13:RP99149. [PMID: 39902803 PMCID: PMC11793870 DOI: 10.7554/elife.99149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025] Open
Abstract
RNAs in circulation carry sequence-specific regulatory information between cells in plant, animal, and host-pathogen systems. Such RNA can cross generational boundaries, as evidenced by somatic double-stranded RNA (dsRNA) in the nematode Caenorhabditis elegans silencing genes of matching sequence in progeny. Here we dissect the intergenerational path taken by dsRNA from parental circulation and discover that cytosolic import through the dsRNA importer SID-1 in the parental germline and/or developing progeny varies with developmental time and dsRNA substrates. Loss of SID-1 enhances initiation of heritable RNA silencing within the germline and causes changes in the expression of the sid-1-dependent gene sdg-1 that last for more than 100 generations after restoration of SID-1. The SDG-1 protein is enriched in perinuclear germ granules required for heritable RNA silencing but is expressed from a retrotransposon targeted by such silencing. This auto-inhibitory loop suggests how retrotransposons could persist by hosting genes that regulate their own silencing.
Collapse
Affiliation(s)
| | - Aishwarya Sathya
- Department of Cell Biology and Molecular Genetics, University of MarylandCollege ParkUnited States
| | - Andrew L Yi
- Department of Cell Biology and Molecular Genetics, University of MarylandCollege ParkUnited States
| | - Winnie M Chan
- Department of Cell Biology and Molecular Genetics, University of MarylandCollege ParkUnited States
| | - Julia A Marre
- Department of Cell Biology and Molecular Genetics, University of MarylandCollege ParkUnited States
| | - Antony M Jose
- Department of Cell Biology and Molecular Genetics, University of MarylandCollege ParkUnited States
| |
Collapse
|
4
|
Sung JY, Lim GE, Goo J, Jung KJ, Chung JM, Jung HS, Kim YN, Shim J. TMEM39A and TMEM131 facilitate bulk transport of ECM proteins through large COPII vesicle formation. J Genet Genomics 2025; 52:189-203. [PMID: 39521045 DOI: 10.1016/j.jgg.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
The growth of Caenorhabditis elegans involves multiple molting processes, during which old cuticles are shed and new cuticles are rapidly formed. This process requires the regulated bulk secretion of cuticle components. The transmembrane protein-39 (TMEM-39) mutant exhibits distinct dumpy and ruptured phenotypes characterized by notably thin cuticles. TMEM-39 primarily co-localizes with the coat protein II complex (COPII) in large vesicles rather than small COPII vesicles. These TMEM-39-associated large vesicles (TMEM-39-LVs) form robustly during the molting period and co-localize with various extracellular matrix components, including BLI-1 collagen, BLI-3 dual oxidase, and carboxypeptidases. Through immunoprecipitation using TMEM39A-FLAG and proteomics analysis in human sarcoma cells, we identify TMEM39A-associated proteins, including TMEM131. Knockdown of TMEM131 results in reduced TMEM39A-LV formation and collagen secretion in both C. elegans and human sarcoma cells, indicating a cooperative role between TMEM39A and TMEM131 in the secretion of extracellular components through the formation of large COPII vesicles. Given the conservation of TMEM39A and its associated proteins between C. elegans and humans, TMEM39A-LVs may represent a fundamental machinery for rapid and extensive secretion across metazoans.
Collapse
Affiliation(s)
- Jee Young Sung
- Cancer Metastasis Branch, Research Institute, National Cancer Center, 323 Ilsan-ro, Goyang-si, Gyeonggi-do, 10408, Republic of Korea
| | - Ga-Eun Lim
- Cancer Metastasis Branch, Research Institute, National Cancer Center, 323 Ilsan-ro, Goyang-si, Gyeonggi-do, 10408, Republic of Korea
| | - Jarim Goo
- Cancer Metastasis Branch, Research Institute, National Cancer Center, 323 Ilsan-ro, Goyang-si, Gyeonggi-do, 10408, Republic of Korea
| | - Kyung Jin Jung
- Experimental Clinical Research Center, Biomedical Research Institute, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, 13620, Republic of Korea
| | - Jeong Min Chung
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Kangwon-do, 24341, Republic of Korea
| | - Hyun Suk Jung
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Kangwon-do, 24341, Republic of Korea
| | - Yong-Nyun Kim
- Cancer Metastasis Branch, Research Institute, National Cancer Center, 323 Ilsan-ro, Goyang-si, Gyeonggi-do, 10408, Republic of Korea.
| | - Jaegal Shim
- Cancer Metastasis Branch, Research Institute, National Cancer Center, 323 Ilsan-ro, Goyang-si, Gyeonggi-do, 10408, Republic of Korea.
| |
Collapse
|
5
|
Lee HJ, Liang J, Chaudhary S, Moon S, Yu Z, Wu T, Liu H, Choi MK, Zhang Y, Lu H. Automated cell annotation in multi-cell images using an improved CRF_ID algorithm. eLife 2025; 12:RP89050. [PMID: 39853076 PMCID: PMC11759411 DOI: 10.7554/elife.89050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2025] Open
Abstract
Cell identification is an important yet difficult process in data analysis of biological images. Previously, we developed an automated cell identification method called CRF_ID and demonstrated its high performance in Caenorhabditis elegans whole-brain images (Chaudhary et al., 2021). However, because the method was optimized for whole-brain imaging, comparable performance could not be guaranteed for application in commonly used C. elegans multi-cell images that display a subpopulation of cells. Here, we present an advancement, CRF_ID 2.0, that expands the generalizability of the method to multi-cell imaging beyond whole-brain imaging. To illustrate the application of the advance, we show the characterization of CRF_ID 2.0 in multi-cell imaging and cell-specific gene expression analysis in C. elegans. This work demonstrates that high-accuracy automated cell annotation in multi-cell imaging can expedite cell identification and reduce its subjectivity in C. elegans and potentially other biological images of various origins.
Collapse
Affiliation(s)
- Hyun Jee Lee
- School of Chemical & Biomolecular Engineering, Georgia Institute of TechnologyAtlantaUnited States
| | - Jingting Liang
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Shivesh Chaudhary
- School of Chemical & Biomolecular Engineering, Georgia Institute of TechnologyAtlantaUnited States
| | - Sihoon Moon
- School of Chemical & Biomolecular Engineering, Georgia Institute of TechnologyAtlantaUnited States
| | - Zikai Yu
- Interdisciplinary BioEngineering Program, Georgia Institute of TechnologyAtlantaUnited States
| | - Taihong Wu
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - He Liu
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Myung-Kyu Choi
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Yun Zhang
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
- Center for Brain Science, Harvard UniversityCambridgeUnited States
| | - Hang Lu
- School of Chemical & Biomolecular Engineering, Georgia Institute of TechnologyAtlantaUnited States
- Interdisciplinary BioEngineering Program, Georgia Institute of TechnologyAtlantaUnited States
| |
Collapse
|
6
|
Jin EJ, Qi YB, Chisholm AD, Jin Y. The BEN domain protein LIN-14 coordinates neuromuscular positioning during epidermal maturation. iScience 2025; 28:111577. [PMID: 39817198 PMCID: PMC11732705 DOI: 10.1016/j.isci.2024.111577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/01/2024] [Accepted: 12/09/2024] [Indexed: 01/18/2025] Open
Abstract
Development and function of an organism depend on coordinated inter-tissue interaction. How such interactions are maintained during tissue renewal and reorganization remains poorly understood. Here, we find that Caenorhabditis elegans BEN domain transcription factor LIN-14 is required in epidermis for maintaining the position of motor neurons and muscles during developmental tissue reorganization. lin-14 loss of function (lf) mutants display highly penetrant ventral neuromuscular mispositioning. These defects arise post-embryonically during first larval (L1) stage as the maturing epidermis replaces the embryonic ventral epidermis. Tissue-specific and temporally controlled depletion experiments indicate LIN-14 acts within the epidermis for ventral neuromuscular positioning. lin-14(lf) mutants show defects in formation of epidermis-muscle attachment complex hemidesmosomes in the maturing ventral epidermis, leading to detachment of muscles and motor neurons as well as movement defects. Our findings reveal a cell non-autonomous role for LIN-14 in coordinating inter-tissue interaction and neuromuscular positioning during epidermal maturation.
Collapse
Affiliation(s)
- Eugene Jennifer Jin
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yingchuan Billy Qi
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Andrew D. Chisholm
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yishi Jin
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
- Kavli Institute of Brain and Mind, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
7
|
Qu Z, Zhang L, Yin X, Dai F, Huang W, Zhang Y, Ran D, Zheng S. Male sex determination maintains proteostasis and extends lifespan of daf-18/PTEN deficient C. elegans. EMBO Rep 2025:10.1038/s44319-025-00368-x. [PMID: 39820856 DOI: 10.1038/s44319-025-00368-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/24/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025] Open
Abstract
Although females typically have a survival advantage, those with PTEN functional abnormalities face a higher risk of developing tumors than males. However, the differences in how each sex responds to PTEN dysfunction have rarely been studied. We use Caenorhabditis elegans to investigate how male and hermaphrodite worms respond to dysfunction of the PTEN homolog daf-18. Our study reveals that male worms can counterbalance the negative effects of daf-18 deficiency, resulting in longer adult lifespan. The survival advantage depends on the loss of DAF-18 protein phosphatase activity, while its lipid phosphatase activity is dispensable. The deficiency in DAF-18 protein phosphatase activity leads to the failure of dephosphorylation of the endoplasmic reticulum membrane protein C18E9.2/SEC62, causing increased levels of unfolded and aggregated proteins in hermaphrodites. In contrast, males maintain proteostasis through a UNC-23/NEF-mediated protein ubiquitination and degradation process, providing them with a survival advantage. We find that sex determination is a key factor in regulating the differential expression of unc-23 between sexes in response to daf-18 loss. These findings highlight the unique role of the male sex determination pathway in regulating protein degradation.
Collapse
Affiliation(s)
- Zhi Qu
- The Zhongzhou Laboratory for Integrative Biology, Henan University, 450000, Zhengzhou, Henan, China
- School of Nursing and Health, Henan University, 475004, Kaifeng, China
| | - Lu Zhang
- School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Xue Yin
- School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Fangzhou Dai
- School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Wei Huang
- School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Yutong Zhang
- School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Dongyang Ran
- School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Shanqing Zheng
- The Zhongzhou Laboratory for Integrative Biology, Henan University, 450000, Zhengzhou, Henan, China.
- School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China.
- Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Medical School of Henan University, 475004, Kaifeng, China.
| |
Collapse
|
8
|
Calarco JA, Taylor SR, Miller DM. Detecting gene expression in Caenorhabditis elegans. Genetics 2025; 229:1-108. [PMID: 39693264 DOI: 10.1093/genetics/iyae167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 09/30/2024] [Indexed: 12/20/2024] Open
Abstract
Reliable methods for detecting and analyzing gene expression are necessary tools for understanding development and investigating biological responses to genetic and environmental perturbation. With its fully sequenced genome, invariant cell lineage, transparent body, wiring diagram, detailed anatomy, and wide array of genetic tools, Caenorhabditis elegans is an exceptionally useful model organism for linking gene expression to cellular phenotypes. The development of new techniques in recent years has greatly expanded our ability to detect gene expression at high resolution. Here, we provide an overview of gene expression methods for C. elegans, including techniques for detecting transcripts and proteins in situ, bulk RNA sequencing of whole worms and specific tissues and cells, single-cell RNA sequencing, and high-throughput proteomics. We discuss important considerations for choosing among these techniques and provide an overview of publicly available online resources for gene expression data.
Collapse
Affiliation(s)
- John A Calarco
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada, M5S 3G5
| | - Seth R Taylor
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| | - David M Miller
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, USA
- Neuroscience Program, Vanderbilt University, Nashville, TN 37240, USA
| |
Collapse
|
9
|
Biswas K, Moore C, Rogers H, Wani KA, Pukkila-Worley R, Higgins DP, Walker AK, Rand JB, Francis MM. Transcriptional responses to prolonged oxidative stress require cholinergic activation of G-protein-coupled receptor signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.628021. [PMID: 39829818 PMCID: PMC11741395 DOI: 10.1101/2025.01.06.628021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Organisms have evolved protective strategies that are geared toward limiting cellular damage and enhancing organismal survival in the face of environmental stresses, but how these protective mechanisms are coordinated remains unclear. Here, we define a requirement for neural activity in mobilizing the antioxidant defenses of the nematode Caenorhabditis elegans both during prolonged oxidative stress and prior to its onset. We show that acetylcholine-deficient mutants are particularly vulnerable to prolonged oxidative stress. We find that prolonged oxidative stress mobilizes a broad transcriptional response which is strongly dependent on both cholinergic signaling and activation of the muscarinic G-protein acetylcholine coupled receptor (mAChR) GAR-3. Gene enrichment analysis revealed a lack of upregulation of proteasomal proteolysis machinery in both cholinergic-deficient and gar-3 mAChR mutants, suggesting that muscarinic activation is critical for stress-responsive upregulation of protein degradation pathways. Further, we find that GAR-3 overexpression in cholinergic motor neurons prolongs survival during prolonged oxidative stress. Our studies demonstrate neuronal modulation of antioxidant defenses through cholinergic activation of G protein-coupled receptor signaling pathways, defining new potential links between cholinergic signaling, oxidative damage, and neurodegenerative disease.
Collapse
|
10
|
Daniele T, Cury J, Morin MC, Ahier A, Isaia D, Jarriault S. Essential and dual effects of Notch activity on a natural transdifferentiation event. Nat Commun 2025; 16:75. [PMID: 39746948 PMCID: PMC11697417 DOI: 10.1038/s41467-024-55286-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025] Open
Abstract
Cell identity can be reprogrammed, naturally or experimentally, albeit with low frequency. Why some cells, but not their neighbours, undergo a cell identity conversion remains unclear. We find that Notch signalling plays a key role to promote natural transdifferentiation in C. elegans hermaphrodites. Endogenous Notch signalling endows a cell with the competence to transdifferentiate by promoting plasticity factors expression (hlh-16/Olig and sem-4/Sall). Strikingly, ectopic Notch can trigger additional transdifferentiation in vivo. However, Notch signalling can both promote and block transdifferentiation depending on its activation timing. Notch only promotes transdifferentiation during an early precise window of opportunity and signal duration must be tightly controlled in time. Our findings emphasise the importance of temporality and dynamics of the underlying molecular events preceding the initiation of natural cell reprogramming. Finally, our results support a model where both an extrinsic signal and the intrinsic cellular context combine to empower a cell with the competence to transdifferentiate.
Collapse
Affiliation(s)
- Thomas Daniele
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM U1298, Université de Strasbourg, Illkirch, France
- Vertex Pharmaceuticals (CH) GmbH, Zug, Switzerland
| | - Jeanne Cury
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM U1298, Université de Strasbourg, Illkirch, France
| | - Marie-Charlotte Morin
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM U1298, Université de Strasbourg, Illkirch, France
| | - Arnaud Ahier
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM U1298, Université de Strasbourg, Illkirch, France
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Davide Isaia
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM U1298, Université de Strasbourg, Illkirch, France
- Skyhawk Therapeutics, Basel, Switzerland
| | - Sophie Jarriault
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM U1298, Université de Strasbourg, Illkirch, France.
| |
Collapse
|
11
|
Donato A, Ritchie FK, Lu L, Wadia M, Martinez-Marmol R, Kaulich E, Sankorrakul K, Lu H, Coakley S, Coulson EJ, Hilliard MA. OSP-1 protects neurons from autophagic cell death induced by acute oxidative stress. Nat Commun 2025; 16:300. [PMID: 39746999 PMCID: PMC11696186 DOI: 10.1038/s41467-024-55105-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 11/21/2024] [Indexed: 01/04/2025] Open
Abstract
Oxidative stress, caused by the accumulation of reactive oxygen species (ROS), is a pathological factor in several incurable neurodegenerative conditions as well as in stroke. However, our knowledge of the genetic elements that can be manipulated to protect neurons from oxidative stress-induced cell death is still very limited. Here, using Caenorhabditis elegans as a model system, combined with the optogenetic tool KillerRed to spatially and temporally control ROS generation, we identify a previously uncharacterized gene, oxidative stress protective 1 (osp-1), that protects C. elegans neurons from oxidative damage. Using rodent and human cell cultures, we also show that the protective effect of OSP-1 extends to mammalian cells. Moreover, we demonstrate that OSP-1 functions in a strictly cell-autonomous fashion, and that it localizes to the endoplasmic reticulum (ER) where it has an ER-remodeling function. Finally, we present evidence suggesting that OSP-1 may exert its neuroprotective function by influencing autophagy. Our results point to a potential role of OSP-1 in modulating autophagy, and suggest that overactivation of this cellular process could contribute to neuronal death triggered by oxidative damage.
Collapse
Affiliation(s)
- Alessandra Donato
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Fiona K Ritchie
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Lachlan Lu
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Mehershad Wadia
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Ramon Martinez-Marmol
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Eva Kaulich
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Kornraviya Sankorrakul
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Hang Lu
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Sean Coakley
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Elizabeth J Coulson
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Massimo A Hilliard
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
12
|
Montresor S, Pigazzini ML, Baskaran S, Sleiman M, Adhikari G, Basilicata L, Secker L, Jacob N, Ehlert Y, Kelkar A, Kalsi GK, Kulkarni N, Spellerberg P, Kirstein J. HSP110 is a modulator of amyloid beta (Aβ) aggregation and proteotoxicity. J Neurochem 2025; 169:e16214. [PMID: 39180255 PMCID: PMC11657929 DOI: 10.1111/jnc.16214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 07/01/2024] [Accepted: 08/13/2024] [Indexed: 08/26/2024]
Abstract
Chaperones safeguard protein homeostasis by promoting folding and preventing aggregation. HSP110 is a cytosolic chaperone that functions as a nucleotide exchange factor for the HSP70 cycle. Together with HSP70 and a J-domain protein (JDP), HSP110 maintains protein folding and resolubilizes aggregates. Interestingly, HSP110 is vital for the HSP70/110/JDP-mediated disaggregation of amyloidogenic proteins implicated in neurodegenerative diseases (i.e., α-synuclein, HTT, and tau). However, despite its abundance, HSP110 remains still an enigmatic chaperone, and its functional spectrum is not very well understood. Of note, the disaggregation activity of neurodegenerative disease-associated amyloid fibrils showed both beneficial and detrimental outcomes in vivo. To gain a more comprehensive understanding of the chaperone HSP110 in vivo, we analyzed its role in neuronal proteostasis and neurodegeneration in C. elegans. Specifically, we investigated the role of HSP110 in the regulation of amyloid beta peptide (Aβ) aggregation using an established Aβ-C. elegans model that mimics Alzheimer's disease pathology. We generated a novel C. elegans model that over-expresses hsp-110 pan-neuronally, and we also depleted hsp-110 by RNAi-mediated knockdown. We assessed Aβ aggregation in vivo and in situ by fluorescence lifetime imaging. We found that hsp-110 over-expression exacerbated Aβ aggregation and appeared to reduce the conformational variability of the Aβ aggregates, whereas hsp-110 depletion reduced aggregation more significantly in the IL2 neurons, which marked the onset of Aβ aggregation. HSP-110 also plays a central role in growth and fertility as its over-expression compromises nematode physiology. In addition, we found that HSP-110 modulation affects the autophagy pathway. While hsp-110 over-expression impairs the autophagic flux, a depletion enhances it. Thus, HSP-110 regulates multiple nodes of the proteostasis network to control amyloid protein aggregation, disaggregation, and autophagic clearance.
Collapse
Affiliation(s)
| | - Maria Lucia Pigazzini
- Leibniz Institute for Molecular PharmacologyBerlinGermany
- Present address:
EMBL HeidelbergMeyerhofstrasse 169117HeidelbergGermany
| | | | - Mira Sleiman
- Department of Cell BiologyUniversity of BremenBremenGermany
- Leibniz Institute on Aging—Fritz‐Lipmann‐InstituteJenaGermany
| | | | | | - Luca Secker
- Department of Cell BiologyUniversity of BremenBremenGermany
| | - Natascha Jacob
- Department of Cell BiologyUniversity of BremenBremenGermany
| | - Yara Ehlert
- Department of Cell BiologyUniversity of BremenBremenGermany
| | | | | | - Niraj Kulkarni
- Department of Cell BiologyUniversity of BremenBremenGermany
| | | | - Janine Kirstein
- Leibniz Institute on Aging—Fritz‐Lipmann‐InstituteJenaGermany
- Friedrich‐Schiller‐Universität, Institute for Biochemistry & BiophysicsJenaGermany
| |
Collapse
|
13
|
Bayansan O, Bhan P, Chang CY, Barmaver SN, Shen CP, Wagner OI. UNC-10/SYD-2 links kinesin-3 to RAB-3-containing vesicles in the absence of the motor's PH domain. Neurobiol Dis 2025; 204:106766. [PMID: 39662532 DOI: 10.1016/j.nbd.2024.106766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 11/20/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024] Open
Abstract
Kinesin-3 KIF1A (UNC-104 in C. elegans) is the major axonal transporter of synaptic vesicles and mutations in this molecular motor are linked to KIF1A-associated neurological disorders (KAND), encompassing Charcot-Marie-Tooth disease, amyotrophic lateral sclerosis and hereditary spastic paraplegia. UNC-104 binds to lipid bilayers of synaptic vesicles via its C-terminal PH (pleckstrin homology) domain. Since this interaction is relatively weak and non-specific, we hypothesize that other, more specific, interaction schemes exist. From the literature, it is evident that UNC-104 regulator SYD-2 interacts with UNC-10 and that UNC-10 itself interacts with RAB-3 bound to synaptic vesicles. RT-PCR and Western blot experiments expose genetic relationships between unc-10 and syd-2, but not between unc-10 and rab-3. Also, neither unc-10 nor rab-3 affects UNC-104 expression. However, co-immunoprecipitation and bimolecular fluorescence complementation (BiFC) assays reveal functional interactions between UNC-104, SYD-2, UNC-10 and RAB-3. Though both SNB-1 and RAB-3 are actively transported by UNC-104, motility of RAB-3 is facilitated in the presence of SYD-2 and UNC-10. Deletion of UNC-104's PH domain did not affect UNC-104/RAB-3 colocalization, but significantly affected UNC-104/SNB-1 colocalization. Similarly, motility of RAB-3-labeled vesicles is only slightly altered in nematodes carrying a point mutation in the PH domain, whereas movement of SNB-1 is significantly reduced in this mutant. Western blots from purified fractions of synaptic vesicles reveal strong reduction of UNC-104 in rab-3/unc-10 double mutants. Our findings suggest that the UNC-10/SYD-2 complex acts as a functional linker to connect UNC-104 to RAB-3-containing vesicles. Thus, this linker complex contributes to the specificity of motor/cargo interactions.
Collapse
Affiliation(s)
- Odvogmed Bayansan
- National Tsing Hua University, Institute of Molecular and Cellular Biology, Department of Life Science, Hsinchu 30013, Taiwan, ROC
| | - Prerana Bhan
- National Tsing Hua University, Institute of Molecular and Cellular Biology, Department of Life Science, Hsinchu 30013, Taiwan, ROC
| | - Chien-Yu Chang
- National Tsing Hua University, Institute of Molecular and Cellular Biology, Department of Life Science, Hsinchu 30013, Taiwan, ROC
| | - Syed Nooruzuha Barmaver
- National Tsing Hua University, Institute of Molecular and Cellular Biology, Department of Life Science, Hsinchu 30013, Taiwan, ROC
| | - Che-Piao Shen
- National Tsing Hua University, Institute of Molecular and Cellular Biology, Department of Life Science, Hsinchu 30013, Taiwan, ROC
| | - Oliver Ingvar Wagner
- National Tsing Hua University, Institute of Molecular and Cellular Biology, Department of Life Science, Hsinchu 30013, Taiwan, ROC.
| |
Collapse
|
14
|
Khan H, Huang X, Raj V, Wang H. A versatile site-directed gene trap strategy to manipulate gene activity and control gene expression in Caenorhabditis elegans. PLoS Genet 2025; 21:e1011541. [PMID: 39841730 PMCID: PMC11753634 DOI: 10.1371/journal.pgen.1011541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/15/2024] [Indexed: 01/24/2025] Open
Abstract
The ability to manipulate gene activity and control transgene expression is essential to study gene function. While several genetic tools for modifying genes or controlling expression separately are available for Caenorhabditis elegans, there are no genetic approaches to generate mutations that simultaneously disrupt gene function and provide genetic access to the cells expressing the disrupted gene. To achieve this, we developed a versatile gene trap strategy based on cGAL, a GAL4-UAS bipartite expression system for C. elegans. We designed a cGAL gene trap cassette and used CRISPR/Cas9 to insert it into the target gene, creating a bicistronic operon that simultaneously expresses a truncated endogenous protein and the cGAL driver in the cells expressing the target gene. We demonstrate that our cGAL gene trap strategy robustly generated loss-of-function alleles. Combining the cGAL gene trap lines with different UAS effector strains allowed us to rescue the loss-of-function phenotype, observe the gene expression pattern, and manipulate cell activity spatiotemporally. We show that, by recombinase-mediated cassette exchange (RMCE) via microinjection or genetic crossing, the cGAL gene trap lines can be further engineered in vivo to easily swap cGAL with other bipartite expression systems' drivers, including QF/QF2, Tet-On/Tet-Off, and LexA, to generate new gene trap lines with different drivers at the same genomic locus. These drivers can be combined with their corresponding effectors for orthogonal transgenic control. Thus, our cGAL-based gene trap is versatile and represents a powerful genetic tool for gene function analysis in C. elegans, which will ultimately provide new insights into how genes in the genome control the biology of an organism.
Collapse
Affiliation(s)
- Haania Khan
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Xinyu Huang
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Genetics Training Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Vishnu Raj
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Han Wang
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Genetics Training Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
15
|
Pender CL, Dishart JG, Gildea HK, Nauta KM, Page EM, Siddiqi TF, Cheung SS, Joe L, Burton NO, Dillin A. Perception of a pathogenic signature initiates intergenerational protection. Cell 2024:S0092-8674(24)01342-4. [PMID: 39721586 DOI: 10.1016/j.cell.2024.11.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 10/10/2024] [Accepted: 11/18/2024] [Indexed: 12/28/2024]
Abstract
Transmission of immune responses from one generation to the next represents a powerful adaptive mechanism to protect an organism's descendants. Parental infection by the natural C. elegans pathogen Pseudomonas vranovensis induces a protective response in progeny, but the bacterial cues and intergenerational signal driving this response were previously unknown. Here, we find that animals activate a protective stress response program upon exposure to P. vranovensis-derived cyanide and that a metabolic byproduct of cyanide detoxification, β-cyanoalanine, acts as an intergenerational signal to protect progeny from infection. Remarkably, this mechanism does not require direct parental infection; rather, exposure to pathogen-derived volatiles is sufficient to enhance the survival of the next generation, indicating that parental surveillance of environmental cues can activate a protective intergenerational response. Therefore, the mere perception of a pathogen-derived toxin, in this case cyanide, can protect an animal's progeny from future pathogenic challenges.
Collapse
Affiliation(s)
- Corinne L Pender
- Department of Molecular & Cellular Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Julian G Dishart
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Holly K Gildea
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kelsie M Nauta
- Department of Metabolism and Nutritional Programming, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Emily M Page
- Department of Molecular & Cellular Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Talha F Siddiqi
- Department of Molecular & Cellular Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Shannon S Cheung
- Department of Molecular & Cellular Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Larry Joe
- Department of Molecular & Cellular Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Nicholas O Burton
- Department of Metabolism and Nutritional Programming, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Andrew Dillin
- Department of Molecular & Cellular Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
16
|
Kimmich MJ, Geary MA, Mi-Mi L, Votra SD, Pellenz CD, Sundaramurthy S, Pruyne D. The sole essential low molecular weight tropomyosin isoform of Caenorhabditis elegans is essential for pharyngeal muscle function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.13.628433. [PMID: 39764053 PMCID: PMC11702560 DOI: 10.1101/2024.12.13.628433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Tropomyosin is an actin-binding protein that plays roles ranging from regulating muscle contraction to controlling cytokinesis and cell migration. The simple nematode Caenorhabditis elegans provides a useful model for studying the core functions of tropomyosin in an animal, having a relatively simple anatomy, and a single tropomyosin gene, lev-11, that produces seven isoforms. Three higher molecular weight isoforms (LEV-11A, D, O) regulate contraction of body wall and other muscles, but comparatively less is known of the functions of four lower molecular weight isoforms (LEV-11C, E, T, U). We demonstrate here C. elegans can survive with a single low molecular weight isoform, LEV-11E. Mutants disrupted for LEV-11E die as young larvae, whereas mutants disrupted for all other short isoforms are viable with no overt phenotype. Vertebrate low molecular weight tropomyosins are often considered "nonmuscle" isoforms, but we find LEV-11E localizes to sarcomeric thin filaments in pharyngeal muscle, and co-precipitates from worm extracts with the formin FHOD-1, which is also associated with thin filaments in pharyngeal muscle. Pharyngeal sarcomere organization is grossly normal in larvae lacking LEV-11E, indicating the tropomyosin is not required to stabilize thin filaments, but pharyngeal pumping is absent, suggesting LEV-11E regulates actomyosin activity similar to higher molecular weight sarcomeric tropomyosin isoforms.
Collapse
Affiliation(s)
- Michael J Kimmich
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 766 Irving Avenue, Syracuse, NY 13210
| | - Meaghan A Geary
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 766 Irving Avenue, Syracuse, NY 13210
| | - Lei Mi-Mi
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 766 Irving Avenue, Syracuse, NY 13210
| | - SarahBeth D Votra
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 766 Irving Avenue, Syracuse, NY 13210
| | - Christopher D Pellenz
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 766 Irving Avenue, Syracuse, NY 13210
| | - Sumana Sundaramurthy
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 766 Irving Avenue, Syracuse, NY 13210
| | - David Pruyne
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 766 Irving Avenue, Syracuse, NY 13210
| |
Collapse
|
17
|
Dinneen E, Silva-García CG. Universal Single Copy Knock-In System in Caenorhabditis elegans : One Plasmid to Target All Chromosomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.06.627295. [PMID: 39713286 PMCID: PMC11661065 DOI: 10.1101/2024.12.06.627295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Successful transgenesis in model organisms has dramatically helped us understand gene function, regulation, genetic networks, and potential applications. Here, we introduce the universal single-copy knock-in system (Universal SKI System or U-SKI), designed for inserting any transgene by CRISPR/Cas9 in the Caenorhabditis elegans genome. The Universal SKI System takes advantage of a plasmid (pSKI), which can also be used for extrachromosomal arrays, to facilitate the insertion of a transgene at specific safe harbor loci on each autosomal chromosome. The pSKI plasmid contains multiple restriction sites for easy cloning and serves as a CRISPR/Cas9-based insertion repair template because it has two synthetic and long homology arms that recombine with the SKI cassettes. This system also uses a single crRNA guide, which acts as a Co-CRISPR enrichment marker. Overall, the Universal SKI System is highly flexible; with the same Universal SKI cassette on each autosome, researchers can select the insertion site and streamline tracking while reducing the complexity of expressing single-copy transgenes in C. elegans .
Collapse
|
18
|
Jia Q, Young D, Zhang Q, Sieburth D. Endogenous hydrogen peroxide positively regulates secretion of a gut-derived peptide in neuroendocrine potentiation of the oxidative stress response in Caenorhabditis elegans. eLife 2024; 13:RP97503. [PMID: 39636673 PMCID: PMC11620748 DOI: 10.7554/elife.97503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Abstract
The gut-brain axis mediates bidirectional signaling between the intestine and the nervous system and is critical for organism-wide homeostasis. Here, we report the identification of a peptidergic endocrine circuit in which bidirectional signaling between neurons and the intestine potentiates the activation of the antioxidant response in Caenorhabditis elegans in the intestine. We identify an FMRF-amide-like peptide, FLP-2, whose release from the intestine is necessary and sufficient to activate the intestinal oxidative stress response by promoting the release of the antioxidant FLP-1 neuropeptide from neurons. FLP-2 secretion from the intestine is positively regulated by endogenous hydrogen peroxide (H2O2) produced in the mitochondrial matrix by sod-3/superoxide dismutase, and is negatively regulated by prdx-2/peroxiredoxin, which depletes H2O2 in both the mitochondria and cytosol. H2O2 promotes FLP-2 secretion through the DAG and calcium-dependent protein kinase C family member pkc-2 and by the SNAP25 family member aex-4 in the intestine. Together, our data demonstrate a role for intestinal H2O2 in promoting inter-tissue antioxidant signaling through regulated neuropeptide-like protein exocytosis in a gut-brain axis to activate the oxidative stress response.
Collapse
Affiliation(s)
- Qi Jia
- Development, Stem Cells and Regenerative Medicine PhD program, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
- Neuromedicine Graduate Program, University of Southern CaliforniaLos AngelesUnited States
| | - Drew Young
- Neuroscience Graduate Program, University of Southern CaliforniaLos AngelesUnited States
- Zilkha Neurogenetic Institute, University of Southern CaliforniaLos AngelesUnited States
| | - Qixin Zhang
- Neuromedicine Graduate Program, University of Southern CaliforniaLos AngelesUnited States
- Zilkha Neurogenetic Institute, University of Southern CaliforniaLos AngelesUnited States
| | - Derek Sieburth
- Zilkha Neurogenetic Institute, University of Southern CaliforniaLos AngelesUnited States
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| |
Collapse
|
19
|
Molina-García L, Colinas-Fischer S, Benavides-Laconcha S, Lin L, Clark E, Treloar NJ, García-Minaur-Ortíz B, Butts M, Barnes CP, Barrios A. Conflict during learning reconfigures the neural representation of positive valence and approach behavior. Curr Biol 2024; 34:5470-5483.e7. [PMID: 39547234 DOI: 10.1016/j.cub.2024.10.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/18/2024] [Accepted: 10/08/2024] [Indexed: 11/17/2024]
Abstract
Punishing and rewarding experiences can change the valence of sensory stimuli and guide animal behavior in opposite directions, resulting in avoidance or approach. Often, however, a stimulus is encountered with both positive and negative experiences. How is such conflicting information represented in the brain and resolved into a behavioral decision? We address this question by dissecting a circuit for sexual conditioning in C. elegans. In this learning paradigm, an odor is conditioned with both a punishment (starvation) and a reward (mates), resulting in odor approach. We find that negative and positive experiences are both encoded by the neuropeptide pigment dispersing factor 1 (PDF-1) being released from, and acting on, different neurons. Each experience creates a distinct memory in the circuit for odor processing. This results in the sensorimotor representation of the odor being different in naive and sexually conditioned animals, despite both displaying approach. Our results reveal that the positive valence of a stimulus is not represented in the activity of any single neuron class but flexibly represented within the circuit according to the experiences and predictions associated with the stimulus.
Collapse
Affiliation(s)
- Laura Molina-García
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK.
| | - Susana Colinas-Fischer
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | | | - Lucy Lin
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Emma Clark
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Neythen J Treloar
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | | | - Milly Butts
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Chris P Barnes
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Arantza Barrios
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
20
|
Wen J, Li Y, Qin Y, Yan L, Zhang K, Li A, Wang Z, Yu F, Lai J, Yang W, Liu YU, Qin D, Su H. Lycorine protects motor neurons against TDP-43 proteinopathy-induced degeneration in cross-species models with amyotrophic lateral sclerosis. Pharmacol Res 2024; 210:107518. [PMID: 39603574 DOI: 10.1016/j.phrs.2024.107518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/13/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Aggregation of TAR-DNA binding protein-43 (TDP-43) is a pathological feature present in nearly 97 % cases of amyotrophic lateral sclerosis (ALS), making it an attractive target for pathogenic studies and drug screening. Here, we have performed a high-throughput screening of 1500 compounds from a natural product library and identified that lycorine, a naturally occurring alkaloid, significantly decreases the level of TDP-43A315T in a cellular model. We further demonstrate that lycorine reduces the level of TDP-43A315T both through inhibiting its synthesis and by promoting its degradation by the ubiquitin-proteasome system (UPS). Importantly, treatment with lycorine significantly attenuates TDP-43 proteinopathy and improves functional recovery in TDP-43A315T-expressing Caenorhabditis elegans and mouse models. These findings suggest that lycorine is a promising lead compound that has therapeutic potential for ALS.
Collapse
Affiliation(s)
- Jing Wen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao
| | - Yunhao Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao
| | - Yanzhu Qin
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510799, China
| | - Lingli Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao
| | - Ke Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao
| | - Ang Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao
| | - Ziying Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao
| | - Feng Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao
| | - Jianheng Lai
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510799, China
| | - Wei Yang
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; GuiZhou University Medical College, Guiyang 550025, China
| | - Yong U Liu
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China.
| | - Dajiang Qin
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510799, China.
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao.
| |
Collapse
|
21
|
Schiller NR, Almuhanna SA, Hoppe PE. UNC-82/NUAK kinase is required by myosin A, but not myosin B, to assemble and function in the thick filament arms of C. elegans striated muscle. Cytoskeleton (Hoboken) 2024; 81:753-774. [PMID: 37983932 DOI: 10.1002/cm.21807] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/10/2023] [Accepted: 10/27/2023] [Indexed: 11/22/2023]
Abstract
The mechanisms that ensure proper assembly, activity, and turnover of myosin II filaments are fundamental to a diverse range of cellular processes. In Caenorhabditis elegans striated muscle, thick filaments contain two myosins that are functionally distinct and spatially segregated. Using transgenic double mutants, we demonstrate that the ability of increased myosin A expression to restore muscle structure and movement in myosin B mutants requires UNC-82/NUAK kinase activity. Myosin B function appears unaffected in the kinase-impaired unc-82(e1220) mutant: the recessive antimorphic effects on early assembly of paramyosin and myosin A in this mutant are counteracted by increased myosin B expression and exacerbated by loss of myosin B. Using chimeric myosins and motility assays, we mapped the region of myosin A that requires UNC-82 activity to a 531-amino-acid region of the coiled-coil rod. This region includes the 264-amino-acid Region 1, which is sufficient in chimeric myosins to rescue the essential filament-initiation function of myosin A, as well as two sites that interact with myosin head domains in the Interacting Heads Motif. A specific physical interaction between myosin A and UNC-82::GFP is supported by GFP labeling of ectopic myosin A filaments but not thin filaments. We hypothesize that UNC-82 regulates assembly competence of myosin A during parallel assembly in the filament arms.
Collapse
Affiliation(s)
- NaTasha R Schiller
- Department of Biological Sciences, Western Michigan University, Kalamazoo, Michigan, USA
- Biology Department, Wingate University, Wingate, North Carolina, USA
| | - Sarah A Almuhanna
- Clinical Laboratory Sciences, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Pamela E Hoppe
- Department of Biological Sciences, Western Michigan University, Kalamazoo, Michigan, USA
| |
Collapse
|
22
|
Ono S. Overexpression of Lifeact in the C. elegans body wall muscle causes sarcomere disorganization and embryonic or larval lethality. Front Cell Dev Biol 2024; 12:1504980. [PMID: 39605982 PMCID: PMC11599240 DOI: 10.3389/fcell.2024.1504980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
Lifeact is a short peptide that is widely utilized as a probe for actin filaments in live imaging. However, high concentrations of Lifeact can alter actin filament dynamics and cause artificial modifications to the actin cytoskeleton. Here, I evaluated Caenorhabditis elegans strains expressing Lifeact fused to fluorescent proteins in the body wall muscle. I found that, while low-level expression of Lifeact from a single-copy transgene was appropriate for labeling sarcomeric actin filaments, overexpression of Lifeact from an extrachromosomal array causes severe disorganization of muscle sarcomeres and lethality at an embryonic or larval stage. Therefore, for imaging studies in C. elegans, Lifeact needs to be kept at a low level by proper management of the expression system.
Collapse
Affiliation(s)
- Shoichiro Ono
- Departments of Pathology and Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
23
|
Brown E, Kuszynski S, Akoachere F, Feduccia J, Malatinszky L, Luth ES. Generation of an endogenous auxin inducible degron-tagged SPAS-1/spastin to investigate its targeted depletion in C. elegans neurons. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001328. [PMID: 39583582 PMCID: PMC11582882 DOI: 10.17912/micropub.biology.001328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 11/26/2024]
Abstract
To facilitate investigations of the microtubule severing protein spastin and its specific role in neurons, we aimed to create a C. elegans strain in which the spastin homolog SPAS-1 is visible and can be degraded with spatial and temporal precision. We used CRISPR-Cas9 to fuse an auxin-inducible degron and mScarlet to the endogenous SPAS-1 protein, enabling degradation of SPAS-1 in neurons during desired life stages. DNA sequencing confirmed in-frame insertion with the SPAS-1 N-terminus and fluorescence microscopy revealed endogenous SPAS-1 throughout the CRISPR-edited worms. Auxin treatment in rgef-1::TIR1; mScarlet::AID*::3xFLAG::spas-1 animals reduced mScarlet::SPAS-1 fluorescence in neuronal ganglia.
Collapse
Affiliation(s)
- Emily Brown
- Department of Biology, Simmons University, Boston, Massachusetts, United States
| | - Samantha Kuszynski
- Department of Biology, Simmons University, Boston, Massachusetts, United States
| | - Faith Akoachere
- Department of Biology, Simmons University, Boston, Massachusetts, United States
| | - James Feduccia
- Department of Biology, Simmons University, Boston, Massachusetts, United States
| | - Lili Malatinszky
- Department of Biology, Simmons University, Boston, Massachusetts, United States
| | - Eric S. Luth
- Department of Biology, Simmons University, Boston, Massachusetts, United States
| |
Collapse
|
24
|
Zhebrun A, Ni JZ, Corveleyn L, Roy SG, Sidoli S, Gu SG. Two H3K23 histone methyltransferases, SET-32 and SET-21, function synergistically to promote nuclear RNAi-mediated transgenerational epigenetic inheritance in Caenorhabditis elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.05.622152. [PMID: 39574755 PMCID: PMC11580914 DOI: 10.1101/2024.11.05.622152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Nuclear RNAi in C. elegans induces a set of transgenerationally heritable marks of H3K9me3, H3K23me3, and H3K27me3 at the target genes. The function of H3K23me3 in the nuclear RNAi pathway is largely unknown due to the limited knowledge of H3K23 histone methyltransferase (HMT). In this study we identified SET-21 as a novel H3K23 HMT. By taking combined genetic, biochemical, imaging, and genomic approaches, we found that SET-21 functions synergistically with a previously reported H3K23 HMT SET-32 to deposit H3K23me3 at the native targets of germline nuclear RNAi. We identified a subset of native nuclear RNAi targets that are transcriptionally activated in the set-21;set-32 double mutant. SET-21 and SET-32 are also required for robust transgenerational gene silencing induced by exogenous dsRNA. The set-21;set-32 double mutant strain exhibits an enhanced temperature-sensitive mortal germline phenotype compared to the set-32 single mutant, while the set-21 single mutant animals are fertile. We also found that HRDE-1 and SET-32 are required for cosuppression, a transgene-induced gene silencing phenomenon, in C. elegans germline. Together, these results support a model in which H3K23 HMTs SET-21 and SET-32 function cooperatively to ensure the robustness of germline nuclear RNAi and promotes the germline immortality under the heat stress.
Collapse
Affiliation(s)
- Anna Zhebrun
- Department of Molecular Biology and Biochemistry, Rutgers the State University of New Jersey, 604 Allison Road, Piscataway, NJ, USA, 08854
| | - Julie Z. Ni
- Department of Molecular Biology and Biochemistry, Rutgers the State University of New Jersey, 604 Allison Road, Piscataway, NJ, USA, 08854
| | - Laura Corveleyn
- Laboratory of Pharmaceutical Biotechnology, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, Gent, Belgium, 9000
| | - Siddharth Ghosh Roy
- Department of Molecular Biology and Biochemistry, Rutgers the State University of New Jersey, 604 Allison Road, Piscataway, NJ, USA, 08854
| | - Simone Sidoli
- Department of Chemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, USA10461
| | - Sam G. Gu
- Department of Molecular Biology and Biochemistry, Rutgers the State University of New Jersey, 604 Allison Road, Piscataway, NJ, USA, 08854
| |
Collapse
|
25
|
Turmel-Couture S, Martel PO, Beaulieu L, Lechasseur X, Fotso Dzuna LV, Narbonne P. Bidirectional transfer of a small membrane-impermeable molecule between the Caenorhabditis elegans intestine and germline. J Biol Chem 2024; 300:107963. [PMID: 39510179 DOI: 10.1016/j.jbc.2024.107963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024] Open
Abstract
The extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) is a positive regulator of cell proliferation often upregulated in cancer. Its Caenorhabditis elegans ortholog MPK-1 stimulates germline stem cell (GSC) proliferation nonautonomously from the intestine or somatic gonad. How MPK-1 can perform this task from either of these two tissues however remains unclear. We reasoned that somatic MPK-1 activity could lead to the generation of proproliferative small molecules that could transfer from the intestine and/or somatic gonad to the germline. Here, in support of this hypothesis, we demonstrate that a significant fraction of the small membrane-impermeable fluorescent molecule, 5-carboxyfluorescein, transfers to the germline after its microinjection in the animal's intestine. The larger part of this transfer targets oocytes and requires the germline receptor mediated endocytosis 2 (RME-2) yolk receptor. A minor quantity of the dye is however distributed independently from RME-2 and more widely in the animal, including the distal germline, gonadal sheath, coelomocytes, and hypodermis. We further show that the intestine-to-germline transfer efficiency of this RME-2 independent fraction does not vary together with GSC proliferation rates or MPK-1 activity. Therefore, if germline proliferation was influenced by small membrane-impermeable molecules generated in the intestine, it is unlikely that proliferation would be regulated at the level of molecule transfer rate. Finally, we show that conversely, a similar fraction of germline injected 5-carboxyfluorescein transfers to the intestine, demonstrating transfer bidirectionality. Altogether, our results establish the possibility of an intestine-to-germline signaling axis mediated by small membrane-impermeable molecules that could promote GSC proliferation cell nonautonomously downstream of MPK-1 activity.
Collapse
Affiliation(s)
- Sarah Turmel-Couture
- Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada
| | - Pier-Olivier Martel
- Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada
| | - Lucie Beaulieu
- Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada
| | - Xavier Lechasseur
- Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada
| | | | - Patrick Narbonne
- Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada.
| |
Collapse
|
26
|
Aoki I, Golinelli L, Dunkel E, Bhat S, Bassam E, Beets I, Gottschalk A. Hierarchical regulation of functionally antagonistic neuropeptides expressed in a single neuron pair. Nat Commun 2024; 15:9504. [PMID: 39489735 PMCID: PMC11532408 DOI: 10.1038/s41467-024-53899-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
Neuronal communication involves small-molecule transmitters, gap junctions, and neuropeptides. While neurons often express multiple neuropeptides, our understanding of the coordination of their actions and their mutual interactions remains limited. Here, we demonstrate that two neuropeptides, NLP-10 and FLP-1, released from the same interneuron pair, AVKL/R, exert antagonistic effects on locomotion speed in Caenorhabditis elegans. NLP-10 accelerates locomotion by activating the G protein-coupled receptor NPR-35 on premotor interneurons that promote forward movement. Notably, we establish that NLP-10 is crucial for the aversive response to mechanical and noxious light stimuli. Conversely, AVK-derived FLP-1 slows down locomotion by suppressing the secretion of NLP-10 from AVK, through autocrine feedback via activation of its receptor DMSR-7 in AVK neurons. Our findings suggest that peptidergic autocrine motifs, exemplified by the interaction between NLP-10 and FLP-1, might represent a widespread mechanism in nervous systems across species. These mutual functional interactions among peptidergic co-transmitters could fine-tune brain activity.
Collapse
Affiliation(s)
- Ichiro Aoki
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue-Strasse 15, D-60438, Frankfurt, Germany.
- Department of Biochemistry, Chemistry and Pharmacy, Institute of Biophysical Chemistry, Goethe University, Frankfurt, Germany.
| | | | - Eva Dunkel
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue-Strasse 15, D-60438, Frankfurt, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute of Biophysical Chemistry, Goethe University, Frankfurt, Germany
| | - Shripriya Bhat
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue-Strasse 15, D-60438, Frankfurt, Germany
| | - Erschad Bassam
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue-Strasse 15, D-60438, Frankfurt, Germany
| | - Isabel Beets
- Department of Biology, KU Leuven, Leuven, Belgium
| | - Alexander Gottschalk
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue-Strasse 15, D-60438, Frankfurt, Germany.
- Department of Biochemistry, Chemistry and Pharmacy, Institute of Biophysical Chemistry, Goethe University, Frankfurt, Germany.
| |
Collapse
|
27
|
Yin X, Dai F, Ran D, Zhang Y, Qu Z, Zheng S. Cysteine protease cathepsin B promotes lysosome integrity to extend the lifespan of alternative day fasting worms. Aging Cell 2024; 23:e14286. [PMID: 39046045 PMCID: PMC11561666 DOI: 10.1111/acel.14286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/30/2024] [Accepted: 07/10/2024] [Indexed: 07/25/2024] Open
Abstract
Alternative day fasting (ADF) has been shown to enhance the lifespan of animals. However, human trials evaluating the efficacy of ADF have only recently emerged, presenting challenges due to the extreme nature of this dietary regimen. To better understand the effects of ADF, we investigated its impact using Caenorhabditis elegans as a model organism. Our findings reveal that ADF extends the lifespan of worms nourished on animal-based protein source, while those fed with plant-based protein as the primary protein source do not experience such benefits. Remarkably, initiating ADF during midlife is sufficient to prolong lifespan, whereas implementation during youth results in developmental damage, and in older age, fails to provide additional extension effects. Furthermore, we discovered that midlife ADF up-regulates the expression of two cysteine protease cathepsin B genes, cpr-2 and cpr-5, which preserve lysosomal integrity and enhance its function in digesting aggregated proteins, as well as enhancing lipid metabolism and ameliorating neurodegenerative disease markers and phenomena during aging. This suggests that midlife ADF has long lasting anti-aging effects and may delay the onset of related diseases, specifically in animals consuming animal-based protein source. These findings offer valuable insights into the effects of ADF and provide guidance for future research and potential applications in individuals.
Collapse
Affiliation(s)
- Xue Yin
- School of Basic Medical SciencesHenan UniversityKaifengChina
| | - Fangzhou Dai
- School of Basic Medical SciencesHenan UniversityKaifengChina
| | - Dongyang Ran
- School of Basic Medical SciencesHenan UniversityKaifengChina
| | - Yutong Zhang
- School of Basic Medical SciencesHenan UniversityKaifengChina
| | - Zhi Qu
- School of Nursing and HealthHenan UniversityKaifengChina
| | - Shanqing Zheng
- School of Basic Medical SciencesHenan UniversityKaifengChina
- Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineMedical School of Henan UniversityKaifengChina
- The Zhongzhou Laboratory for Integrative BiologyZhengzhouHenanChina
| |
Collapse
|
28
|
Valera-Alberni M, Yao P, Romero-Sanz S, Lanjuin A, Mair WB. Novel imaging tools to study mitochondrial morphology in Caenorhabditis elegans. Life Sci Alliance 2024; 7:e202402918. [PMID: 39260886 PMCID: PMC11391045 DOI: 10.26508/lsa.202402918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/13/2024] Open
Abstract
Mitochondria exhibit a close interplay between their structure and function. Understanding this intricate relationship requires advanced imaging techniques that can capture the dynamic nature of mitochondria and their impact on cellular processes. However, much of the work on mitochondrial dynamics has been performed in single celled organisms or in vitro cell culture. Here, we introduce novel genetic tools for live imaging of mitochondrial morphology in the nematode Caenorhabditis elegans, addressing a pressing need for advanced techniques in studying organelle dynamics within live intact multicellular organisms. Through a comprehensive analysis, we directly compare our tools with existing methods, demonstrating their advantages for visualizing mitochondrial morphology and contrasting their impact on organismal physiology. We reveal limitations of conventional techniques, whereas showcasing the utility and versatility of our approaches, including endogenous CRISPR tags and ectopic labeling. By providing a guide for selecting the most suitable tools based on experimental goals, our work advances mitochondrial research in C. elegans and enhances the strategic integration of diverse imaging modalities for a holistic understanding of organelle dynamics in living organisms.
Collapse
Affiliation(s)
- Miriam Valera-Alberni
- Department of Molecular Metabolism, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Pallas Yao
- Department of Molecular Metabolism, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Silvia Romero-Sanz
- Department of Molecular Metabolism, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Anne Lanjuin
- Department of Molecular Metabolism, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - William B Mair
- Department of Molecular Metabolism, Harvard TH Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
29
|
Wong C, Jurczak EM, Roy R. Neuronal exosomes transport an miRISC cargo to preserve stem cell integrity during energy stress. Cell Rep 2024; 43:114851. [PMID: 39392750 DOI: 10.1016/j.celrep.2024.114851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 08/19/2024] [Accepted: 09/24/2024] [Indexed: 10/13/2024] Open
Abstract
During periods of nutrient scarcity, many animals undergo germline quiescence to preserve reproductive capacity, and neurons are often necessary for this adaptation. We show here that starvation causes the release of neuronal microRNA (miRNA)/Argonaute-loaded exosomes following AMP kinase-regulated trafficking changes within serotonergic neurons. This neuron-to-germline communication is independent of classical neurotransmission but instead relies on endosome-derived vesicles that carry a pro-quiescent small RNA cargo to modify germline gene expression. Using an miRNA activity sensor, we show that neuronally expressed miRNAs can extinguish the expression of germline mRNA targets in an exosome-dependent manner. Our findings demonstrate how an adaptive neuronal response can change gene expression at a distance by redirecting intracellular trafficking to release neuronal exosomes with specific miRNA cargoes capable of tracking to their appropriate destinations.
Collapse
Affiliation(s)
- Christopher Wong
- Department of Biology, McGill University, Montreal, QC H3A 1B1, Canada
| | - Elena M Jurczak
- Department of Biology, McGill University, Montreal, QC H3A 1B1, Canada
| | - Richard Roy
- Department of Biology, McGill University, Montreal, QC H3A 1B1, Canada.
| |
Collapse
|
30
|
Salazar CJ, Diaz-Balzac CA, Wang Y, Rahman M, Grant BD, Bülow HE. RABR-1, an atypical Rab-related GTPase, cell-nonautonomously restricts somatosensory dendrite branching. Genetics 2024; 228:iyae113. [PMID: 39028768 PMCID: PMC11457943 DOI: 10.1093/genetics/iyae113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/21/2024] Open
Abstract
Neurons are highly polarized cells with dendrites and axons. Dendrites, which receive sensory information or input from other neurons, often display elaborately branched morphologies. While mechanisms that promote dendrite branching have been widely studied, less is known about the mechanisms that restrict branching. Using the nematode Caenorhabditis elegans, we identify rabr-1 (for Rab-related gene 1) as a factor that restricts branching of the elaborately branched dendritic trees of PVD and FLP somatosensory neurons. Animals mutant for rabr-1 show excessively branched dendrites throughout development and into adulthood in areas where the dendrites overlay epidermal tissues. Phylogenetic analyses show that RABR-1 displays similarity to small GTPases of the Rab-type, although based on sequence alone, no clear vertebrate ortholog of RABR-1 can be identified. We find that rabr-1 is expressed and can function in epidermal tissues, suggesting that rabr-1 restricts dendritic branching cell-nonautonomously. Genetic experiments further indicate that for the formation of ectopic branches rabr-1 mutants require the genes of the Menorin pathway, which have been previously shown to mediate dendrite morphogenesis of somatosensory neurons. A translational reporter for RABR-1 reveals a subcellular localization to punctate, perinuclear structures, which correlates with endosomal and autophagosomal markers, but anticorrelates with lysosomal markers suggesting an amphisomal character. Point mutations in rabr-1 analogous to key residues of small GTPases suggest that rabr-1 functions in a GTP-bound form independently of GTPase activity. Taken together, rabr-1 encodes for an atypical small GTPase of the Rab-type that cell-nonautonomously restricts dendritic branching of somatosensory neurons, likely independently of GTPase activity.
Collapse
Affiliation(s)
| | - Carlos A Diaz-Balzac
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Yu Wang
- Department of Molecular Biology and Biochemistry, Rutgers Center for Lipid Research, Rutgers University, Piscataway, NJ 08854, USA
| | - Maisha Rahman
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Barth D Grant
- Department of Molecular Biology and Biochemistry, Rutgers Center for Lipid Research, Rutgers University, Piscataway, NJ 08854, USA
| | - Hannes E Bülow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
31
|
Zhang MG, Seyedolmohadesin M, Mercado SH, Tauffenberger A, Park H, Finnen N, Schroeder FC, Venkatachalam V, Sternberg PW. Sensory integration of food and population density during the diapause exit decision involves insulin-like signaling in Caenorhabditis elegans. Proc Natl Acad Sci U S A 2024; 121:e2405391121. [PMID: 39316052 PMCID: PMC11459166 DOI: 10.1073/pnas.2405391121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 08/20/2024] [Indexed: 09/25/2024] Open
Abstract
Decisions made over long time scales, such as life cycle decisions, require coordinated interplay between sensory perception and sustained gene expression. The Caenorhabditis elegans dauer (or diapause) exit developmental decision requires sensory integration of population density and food availability to induce an all-or-nothing organismal-wide response, but the mechanism by which this occurs remains unknown. Here, we demonstrate how the Amphid Single Cilium J (ASJ) chemosensory neurons, known to be critical for dauer exit, perform sensory integration at both the levels of gene expression and calcium activity. In response to favorable conditions, dauers rapidly produce and secrete the dauer exit-promoting insulin-like peptide INS-6. Expression of ins-6 in the ASJ neurons integrates population density and food level and can reflect decision commitment since dauers committed to exiting have higher ins-6 expression levels than those of noncommitted dauers. Calcium imaging in dauers reveals that the ASJ neurons are activated by food, and this activity is suppressed by pheromone, indicating that sensory integration also occurs at the level of calcium transients. We find that ins-6 expression in the ASJ neurons depends on neuronal activity in the ASJs, cGMP signaling, and the pheromone components ascr#8 and ascr#2. We propose a model in which decision commitment to exit the dauer state involves an autoregulatory feedback loop in the ASJ neurons that promotes high INS-6 production and secretion. These results collectively demonstrate how insulin-like peptide signaling helps animals compute long-term decisions by bridging sensory perception to decision execution.
Collapse
Affiliation(s)
- Mark G. Zhang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | | | - Soraya Hawk Mercado
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - Arnaud Tauffenberger
- Boyce Thompson Institute, Cornell University, Ithaca, NY14853
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY14853
| | - Heenam Park
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - Nerissa Finnen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - Frank C. Schroeder
- Boyce Thompson Institute, Cornell University, Ithaca, NY14853
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY14853
| | | | - Paul W. Sternberg
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| |
Collapse
|
32
|
Xue W, Lei Z, Liu B, Guo H, Yan W, Jin YN, Yu YV. Olfactory dysfunction as an early pathogenic indicator in C. elegans models of Alzheimer's and polyglutamine diseases. Front Aging Neurosci 2024; 16:1462238. [PMID: 39411283 PMCID: PMC11473296 DOI: 10.3389/fnagi.2024.1462238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
Neurodegenerative diseases such as Alzheimer's disease and polyglutamine diseases are characterized by abnormal accumulation of misfolded proteins, leading to neuronal dysfunction and subsequent neuron death. However, there is a lack of studies that integrate molecular, morphological, and functional analyses in neurodegenerative models to fully characterize these time-dependent processes. In this study, we used C. elegans models expressing Aβ1-42 and polyglutamine to investigate early neuronal pathogenic features in olfactory neurons. Both models demonstrated significant reductions in odor sensitivity in AWB and AWC chemosensory neurons as early as day 1 of adulthood, while AWA chemosensory neurons showed no such decline, suggesting cell-type-specific early neuronal dysfunction. At the molecular level, Aβ1-42 or Q40 expression caused age-dependent protein aggregation and morphological changes in neurons. By day 6, both models displayed prominent protein aggregates in neuronal cell bodies and neurites. Notably, AWB neurons in both models showed significantly shortened cilia and increased instances of enlarged cilia as early as day 1 of adulthood. Furthermore, AWC neurons expressing Aβ1-42 displayed calcium signaling defects, with significantly reduced responses to odor stimuli on day 1, further supporting early behavioral dysfunction. In contrast, AWA neuron did not exhibit reduced calcium responses, consistent with the absence of detectable decreases in olfactory sensitivity in these neurons. These findings suggest that decreased calcium signaling and dysfunction in specific sensory neuron subtypes are early indicators of neurodegeneration in C. elegans, occurring prior to the formation of visible protein aggregates. We found that the ER unfolded protein response (UPR) is significantly activated in worms expressing Aβ1-42. Activation of the AMPK pathway alleviates olfactory defects and reduces fibrillar Aβ in these worms. This study underscores the use of C. elegans olfactory neurons as a model to elucidate mechanisms of proteostasis in neurodegenerative diseases and highlights the importance of integrated approaches.
Collapse
Affiliation(s)
- Weikang Xue
- Department of Neurology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Ziyi Lei
- Department of Neurology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Bin Liu
- Department of Neurology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Hanxin Guo
- Department of Neurology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Weiyi Yan
- Department of Neurology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Youngnam N. Jin
- Department of Neurology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Yanxun V. Yu
- Department of Neurology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| |
Collapse
|
33
|
Berndt H, Fuchs S, Kraus-Stojanowic I, Pees B, Gelhaus C, Leippe M. Molecular and functional characterization of ILYS-5, a major invertebrate lysozyme of Caenorhabditis elegans. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 159:105220. [PMID: 38925432 DOI: 10.1016/j.dci.2024.105220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 06/28/2024]
Abstract
To overcome bacterial invasion and infection, animals have evolved various antimicrobial effectors such as antimicrobial peptides and lysozymes. Although C. elegans is exposed to a variety of microbes due to its bacterivorous lifestyle, previous work on the components of its immune system mainly based on the description of transcriptional changes during bacterial challenges. Very few effector components of its immune system have been characterized so far. To investigate the role of lysozymes in terms of antibacterial defense and digestion, we studied a member of the widely neglected family of C. elegans invertebrate lysozymes (ILYS). We focused on the so far virtually undescribed ILYS-5, which we purified from protein extracts of C. elegans tracing its peptidoglycan-degrading activity and localized the tissue expression of the gene in vivo using a translational reporter construct. We recombinantly synthesized ILYS-5 and determined the physicochemical activity optimum and the antibacterial spectrum of a lysozyme from C. elegans for the first time. With an activity optimum at low ionic strength (≤100 mM) and at acidic pH (≤ pH 4.0), ILYS-5 is likely to be involved in killing and digestion of bacteria within acidified phagolysosomes and acidic regions of the gut, presumably secreted by lysosome-like vesicles. This notion is supported by potent activity against various live Gram-positive and Gram-negative bacteria. Notably, members of the natural associated microbiome of C. elegans are substantially less susceptible to ILYS-5. Ablation of the ilys-5 gene resulted in reduction of lifespan and fertility when cultured on the standard food bacterium Escherichia coli OP50, whereas exposure of the ilys-5 knock-out mutant to the host-associated bacterium Pseudomonas lurida MYb11 did not have a clear effect. These findings indicate a role of ILYS-5 in immunity and nutrition and a co-evolved adaptation of host and bacteria to the mutualistic nature of their interaction.
Collapse
Affiliation(s)
- Henry Berndt
- Comparative Immunobiology, Zoological Institute, Kiel University, Kiel, Germany
| | - Silja Fuchs
- Comparative Immunobiology, Zoological Institute, Kiel University, Kiel, Germany
| | | | - Barbara Pees
- Comparative Immunobiology, Zoological Institute, Kiel University, Kiel, Germany
| | - Christoph Gelhaus
- Comparative Immunobiology, Zoological Institute, Kiel University, Kiel, Germany
| | - Matthias Leippe
- Comparative Immunobiology, Zoological Institute, Kiel University, Kiel, Germany.
| |
Collapse
|
34
|
Iguchi R, Kita T, Watanabe T, Chiba K, Niwa S. Characterizing human KIF1Bβ motor activity by single-molecule motility assays and Caenorhabditis elegans genetics. J Cell Sci 2024; 137:jcs261783. [PMID: 39279507 DOI: 10.1242/jcs.261783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 09/05/2024] [Indexed: 09/18/2024] Open
Abstract
The axonal transport of synaptic vesicle precursors relies on KIF1A and UNC-104 ortholog motors. In mammals, KIF1Bβ is also responsible for the axonal transport of synaptic vesicle precursors. Mutations in KIF1A and KIF1Bβ lead to a wide range of neuropathies. Although previous studies have revealed the biochemical, biophysical and cell biological properties of KIF1A, and its defects in neurological disorders, the fundamental properties of KIF1Bβ remain elusive. In this study, we determined the motile parameters of KIF1Bβ through single-molecule motility assays. We found that the C-terminal region of KIF1Bβ has an inhibitory role in motor activity. AlphaFold2 prediction suggests that the C-terminal region blocks the motor domain. Additionally, we established simple methods for testing the axonal transport activity of human KIF1Bβ using Caenorhabditis elegans genetics. Taking advantage of these methods, we demonstrated that these assays enable the detection of reduced KIF1Bβ activities, both in vitro and in vivo, caused by a Charcot-Marie-Tooth disease-associated Q98L mutation.
Collapse
Affiliation(s)
- Rei Iguchi
- Graduate School of Life Sciences , Tohoku University, Katahira 2-1, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Tomoki Kita
- Graduate School of Life Sciences , Tohoku University, Katahira 2-1, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Taisei Watanabe
- Department of Biology, Faculty of Science, Tohoku University, Aramaki-Aoba 6-3, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Kyoko Chiba
- Frontier Research Institute for Interdisciplinary Sciences (FRIS) , Tohoku University, Aramaki-Aoba 6-3, Aoba-ku, Sendai, Miyagi 980-0845, Japan
| | - Shinsuke Niwa
- Graduate School of Life Sciences , Tohoku University, Katahira 2-1, Aoba-ku, Sendai, Miyagi 980-8578, Japan
- Department of Biology, Faculty of Science, Tohoku University, Aramaki-Aoba 6-3, Aoba-ku, Sendai, Miyagi 980-8578, Japan
- Frontier Research Institute for Interdisciplinary Sciences (FRIS) , Tohoku University, Aramaki-Aoba 6-3, Aoba-ku, Sendai, Miyagi 980-0845, Japan
| |
Collapse
|
35
|
Guo A, Wu Q, Yan X, Chen K, Liu Y, Liang D, Yang Y, Luo Q, Xiong M, Yu Y, Fei E, Chen F. Differential roles of lysosomal cholesterol transporters in the development of C. elegans NMJs. Life Sci Alliance 2024; 7:e202402584. [PMID: 39084875 PMCID: PMC11291935 DOI: 10.26508/lsa.202402584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
Cholesterol homeostasis in neurons is critical for synapse formation and maintenance. Neurons with impaired cholesterol uptake undergo progressive synapse loss and eventual degeneration. To investigate the molecular mechanisms of neuronal cholesterol homeostasis and its role during synapse development, we studied motor neurons of Caenorhabditis elegans because these neurons rely on dietary cholesterol. Combining lipidomic analysis, we discovered that NCR-1, a lysosomal cholesterol transporter, promotes cholesterol absorption and synapse development. Loss of ncr-1 causes smaller synapses, and low cholesterol exacerbates the deficits. Moreover, NCR-1 deficiency hinders the increase in synapses under high cholesterol. Unexpectedly, NCR-2, the NCR-1 homolog, increases the use of cholesterol and sphingomyelins and impedes synapse formation. NCR-2 deficiency causes an increase in synapses regardless of cholesterol concentration. Inhibiting the degradation or synthesis of sphingomyelins can induce or suppress the synaptic phenotypes in ncr-2 mutants. Our findings indicate that neuronal cholesterol homeostasis is differentially controlled by two lysosomal cholesterol transporters and highlight the importance of neuronal cholesterol homeostasis in synapse development.
Collapse
Affiliation(s)
- Amin Guo
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qi Wu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xin Yan
- School of Life Sciences, Nanchang University, Nanchang, China
| | - Kanghua Chen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yuxiang Liu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Dingfa Liang
- Queen Mary School of Nanchang University, Jiangxi Medical College, Nanchang, China
| | - Yuxiao Yang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qunfeng Luo
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Mingtao Xiong
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yong Yu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Erkang Fei
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Fei Chen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
36
|
Jia Q, Young D, Zhang Q, Sieburth D. Endogenous hydrogen peroxide positively regulates secretion of a gut-derived peptide in neuroendocrine potentiation of the oxidative stress response in C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587937. [PMID: 39345448 PMCID: PMC11429608 DOI: 10.1101/2024.04.03.587937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The gut-brain axis mediates bidirectional signaling between the intestine and the nervous system and is critical for organism-wide homeostasis. Here we report the identification of a peptidergic endocrine circuit in which bidirectional signaling between neurons and the intestine potentiates the activation of the antioxidant response in C. elegans in the intestine. We identify a FMRF-amide-like peptide, FLP-2, whose release from the intestine is necessary and sufficient to activate the intestinal oxidative stress response by promoting the release of the antioxidant FLP-1 neuropeptide from neurons. FLP-2 secretion from the intestine is positively regulated by endogenous hydrogen peroxide (H2O2) produced in the mitochondrial matrix by sod-3/superoxide dismutase, and is negatively regulated by prdx-2/peroxiredoxin, which depletes H2O2 in both the mitochondria and cytosol. H2O2 promotes FLP-2 secretion through the DAG and calciumdependent protein kinase C family member pkc-2 and by the SNAP25 family member aex-4 in the intestine. Together, our data demonstrate a role for intestinal H2O2 in promoting inter-tissue antioxidant signaling through regulated neuropeptide-like protein exocytosis in a gut-brain axis to activate the oxidative stress response.
Collapse
Affiliation(s)
- Qi Jia
- Development, Stem Cells and Regenerative Medicine PhD program, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
- Neuromedicine Graduate Program, University of Southern California, Los Angeles, CA 90089
| | - Drew Young
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033
| | - Qixin Zhang
- Neuromedicine Graduate Program, University of Southern California, Los Angeles, CA 90089
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033
| | - Derek Sieburth
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| |
Collapse
|
37
|
Adams PE, Thies JL, Sutton JM, Millwood JD, Caldwell GA, Caldwell KA, Fierst JL. Identifying transgene insertions in Caenorhabditis elegans genomes with Oxford Nanopore sequencing. PeerJ 2024; 12:e18100. [PMID: 39285918 PMCID: PMC11404476 DOI: 10.7717/peerj.18100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024] Open
Abstract
Genetically modified organisms are commonly used in disease research and agriculture but the precise genomic alterations underlying transgenic mutations are often unknown. The position and characteristics of transgenes, including the number of independent insertions, influences the expression of both transgenic and wild-type sequences. We used long-read, Oxford Nanopore Technologies (ONT) to sequence and assemble two transgenic strains of Caenorhabditis elegans commonly used in the research of neurodegenerative diseases: BY250 (pPdat-1::GFP) and UA44 (GFP and human α-synuclein), a model for Parkinson's research. After scaffolding to the reference, the final assembled sequences were ∼102 Mb with N50s of 17.9 Mb and 18.0 Mb, respectively, and L90s of six contiguous sequences, representing chromosome-level assemblies. Each of the assembled sequences contained more than 99.2% of the Nematoda BUSCO genes found in the C. elegans reference and 99.5% of the annotated C. elegans reference protein-coding genes. We identified the locations of the transgene insertions and confirmed that all transgene sequences were inserted in intergenic regions, leaving the organismal gene content intact. The transgenic C. elegans genomes presented here will be a valuable resource for Parkinson's research as well as other neurodegenerative diseases. Our work demonstrates that long-read sequencing is a fast, cost-effective way to assemble genome sequences and characterize mutant lines and strains.
Collapse
Affiliation(s)
- Paula E Adams
- Department of Biological Sciences, Auburn University, Auburn, AL, United States of America
- Department of Biological Sciences, University of Alabama - Tuscaloosa, Tuscaloosa, AL, United States of America
| | - Jennifer L Thies
- Department of Biological Sciences, University of Alabama - Tuscaloosa, Tuscaloosa, AL, United States of America
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - John M Sutton
- Department of Biological Sciences, University of Alabama - Tuscaloosa, Tuscaloosa, AL, United States of America
- Absci, Vancouver, WA, United States of America
| | - Joshua D Millwood
- Department of Biological Sciences, University of Alabama - Tuscaloosa, Tuscaloosa, AL, United States of America
- Department of Biological and Environmental Sciences, University of West Alabama, Livingston, AL, United States of America
| | - Guy A Caldwell
- Department of Biological Sciences, University of Alabama - Tuscaloosa, Tuscaloosa, AL, United States of America
| | - Kim A Caldwell
- Department of Biological Sciences, University of Alabama - Tuscaloosa, Tuscaloosa, AL, United States of America
| | - Janna L Fierst
- Department of Biological Sciences, Florida International University, Miami, FL, United States of America
- Biomolecular Sciences Institute, Florida International University, Miami, FL, United States of America
| |
Collapse
|
38
|
Blazie SM, Fortunati D, Zhao Y, Jin Y. C. elegans LIN-66 mediates EIF-3/eIF3-dependent protein translation via a cold-shock domain. Life Sci Alliance 2024; 7:e202402673. [PMID: 38886018 PMCID: PMC11184513 DOI: 10.26508/lsa.202402673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Protein translation initiation is a conserved process involving many proteins acting in concert. The 13 subunit eukaryotic initiation factor 3 (eIF3) complex is essential for assembly of the pre-initiation complex that scans mRNA and positions ribosome at the initiation codon. We previously reported that a gain-of-function (gf) mutation affecting the G subunit of the Caenorhabditis elegans eIF3 complex, eif-3.g(gf), selectively modulates protein translation in the ventral cord cholinergic motor neurons. Here, through unbiased genetic suppressor screening, we identified that the gene lin-66 mediates eif-3.g(gf)-dependent protein translation in motor neurons. LIN-66 is composed largely of low-complexity amino acid sequences with unknown functional domains. We combined bioinformatics analysis with in vivo functional dissection and identified a cold-shock domain in LIN-66 critical for its function. In cholinergic motor neurons, LIN-66 shows a close association with EIF-3.G in the cytoplasm. The low-complexity amino acid sequences of LIN-66 modulate its subcellular pattern. As cold-shock domains function broadly in RNA regulation, we propose that LIN-66 mediates stimulus-dependent protein translation by facilitating the interaction of mRNAs with EIF-3.G.
Collapse
Affiliation(s)
- Stephen M Blazie
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Daniel Fortunati
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Yan Zhao
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Yishi Jin
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
39
|
Sharma N, Au V, Martin K, Edgley ML, Moerman D, Mains PE, Gilleard JS. Multiple UDP glycosyltransferases modulate benzimidazole drug sensitivity in the nematode Caenorhabditis elegans in an additive manner. Int J Parasitol 2024; 54:535-549. [PMID: 38806068 DOI: 10.1016/j.ijpara.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/08/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
Xenobiotic biotransformation is an important modulator of anthelmintic drug potency and a potential mechanism of anthelmintic resistance. Both the free-living nematode Caenorhabditis elegans and the ruminant parasite Haemonchus contortus biotransform benzimidazole drugs by glucose conjugation, likely catalysed by UDP-glycosyltransferase (UGT) enzymes. To identify C. elegans genes involved in benzimidazole drug detoxification, we first used a comparative phylogenetic analysis of UGTs from humans, C. elegans and H. contortus, combined with available RNAseq datasets to identify which of the 63 C. elegans ugt genes are most likely to be involved in benzimidazole drug biotransformation. RNA interference knockdown of 15 prioritized C. elegans genes identified those that sensitized animals to the benzimidazole derivative albendazole (ABZ). Genetic mutations subsequently revealed that loss of ugt-9 and ugt-11 had the strongest effects. The "ugt-9 cluster" includes these genes, together with six other closely related ugts. A CRISPR-Cas-9 deletion that removed seven of the eight ugt-9 cluster genes had greater ABZ sensitivity than the single largest-effect mutation. Furthermore, a double mutant of ugt-22 (which is not a member of the ugt-9 cluster) with the ugt-9 cluster deletion further increased ABZ sensitivity. This additivity of mutant phenotypes suggest that ugt genes act in parallel, which could have several, not mutually exclusive, explanations. ugt mutations have different effects with different benzimidazole derivatives, suggesting that enzymes with different specificities could together more efficiently detoxify drugs. Expression patterns of ugt-9, ugt-11 and ugt-22 gfp reporters differ and so likely act in different tissues which may, at least in part, explain their additive effects on drug potency. Overexpression of ugt-9 alone was sufficient to confer partial ABZ resistance, indicating increasing total UGT activity protects animals. In summary, our results suggest that the multiple UGT enzymes have overlapping but not completely redundant functions in benzimidazole drug detoxification and may represent "druggable" targets to improve benzimidazole drug potency.
Collapse
Affiliation(s)
- Nidhi Sharma
- Host-Parasite Interactions Program, Faculty of Veterinary Medicine, University of Calgary, Alberta, Canada
| | - Vinci Au
- Department of Zoology, Life Sciences Centre, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada
| | - Kiana Martin
- Department of Zoology, Life Sciences Centre, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada
| | - Mark L Edgley
- Department of Zoology, Life Sciences Centre, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada
| | - Don Moerman
- Department of Zoology, Life Sciences Centre, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada
| | - Paul E Mains
- Departments of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - John S Gilleard
- Host-Parasite Interactions Program, Faculty of Veterinary Medicine, University of Calgary, Alberta, Canada.
| |
Collapse
|
40
|
Alirzayeva H, Loureiro R, Koyuncu S, Hommen F, Nabawi Y, Zhang WH, Dao TTP, Wehrmann M, Lee HJ, Vilchez D. ALS-FUS mutations cause abnormal PARylation and histone H1.2 interaction, leading to pathological changes. Cell Rep 2024; 43:114626. [PMID: 39167487 DOI: 10.1016/j.celrep.2024.114626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 05/13/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024] Open
Abstract
The majority of severe early-onset and juvenile cases of amyotrophic lateral sclerosis (ALS) are caused by mutations in the FUS gene, resulting in rapid disease progression. Mutant FUS accumulates within stress granules (SGs), thereby affecting the dynamics of these ribonucleoprotein complexes. Here, we define the interactome of the severe mutant FUSP525L variant in human induced pluripotent stem cell (iPSC)-derived motor neurons. We find increased interaction of FUSP525L with the PARP1 enzyme, promoting poly-ADP-ribosylation (PARylation) and binding of FUS to histone H1.2. Inhibiting PARylation or reducing H1.2 levels alleviates mutant FUS aggregation, SG alterations, and apoptosis in human motor neurons. Conversely, elevated H1.2 levels exacerbate FUS-ALS phenotypes, driven by the internally disordered terminal domains of H1.2. In C. elegans models, knockdown of H1.2 and PARP1 orthologs also decreases FUSP525L aggregation and neurodegeneration, whereas H1.2 overexpression worsens ALS-related changes. Our findings indicate a link between PARylation, H1.2, and FUS with potential therapeutic implications.
Collapse
Affiliation(s)
- Hafiza Alirzayeva
- Institute for Integrated Stress Response Signaling, Faculty of Medicine, University Hospital Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Rute Loureiro
- Institute for Integrated Stress Response Signaling, Faculty of Medicine, University Hospital Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Seda Koyuncu
- Institute for Integrated Stress Response Signaling, Faculty of Medicine, University Hospital Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Franziska Hommen
- Institute for Integrated Stress Response Signaling, Faculty of Medicine, University Hospital Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Yara Nabawi
- Institute for Integrated Stress Response Signaling, Faculty of Medicine, University Hospital Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - William Hongyu Zhang
- Institute for Integrated Stress Response Signaling, Faculty of Medicine, University Hospital Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Thien T P Dao
- Institute for Integrated Stress Response Signaling, Faculty of Medicine, University Hospital Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Markus Wehrmann
- Institute for Integrated Stress Response Signaling, Faculty of Medicine, University Hospital Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Hyun Ju Lee
- Institute for Integrated Stress Response Signaling, Faculty of Medicine, University Hospital Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - David Vilchez
- Institute for Integrated Stress Response Signaling, Faculty of Medicine, University Hospital Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Institute for Genetics, University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
41
|
Chen S, Phillips CM. Silencing of a NRDE-3 transgene in C. elegans germ cells and early embryos is mediated by the RNAi pathway. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001308. [PMID: 39220141 PMCID: PMC11364987 DOI: 10.17912/micropub.biology.001308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Transgenes are highly susceptible to gene silencing in the C. elegans germline. Here, we examine the expression of the nuclear Argonaute protein NRDE-3 , comparing two GFP::NRDE-3 strains, one constructed by bombardment and one by CRISPR. We found that the GFP::NRDE-3 strain constructed by bombardment displays transgene silencing in germline and early embryos and that NRDE-3 expression can be restored in a rde-3 mutant, which disrupts the RNAi pathway. This finding reveals that NRDE-3 is not a soma-specific Argonaute protein and is, in fact, expressed in the proximal germline and early embryos.
Collapse
Affiliation(s)
- Shihui Chen
- Biological Sciences, University of Southern California, Los Angeles, CA, United States
| | - Carolyn M Phillips
- Biological Sciences, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
42
|
Patel R, Bryant AS, Castelletto ML, Walsh B, Akimori D, Hallem EA. The generation of stable transgenic lines in the human-infective nematode Strongyloides stercoralis. G3 (BETHESDA, MD.) 2024; 14:jkae122. [PMID: 38839055 PMCID: PMC11304987 DOI: 10.1093/g3journal/jkae122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/13/2024] [Accepted: 05/23/2024] [Indexed: 06/07/2024]
Abstract
The skin-penetrating gastrointestinal parasitic nematode Strongyloides stercoralis causes strongyloidiasis, which is a neglected tropical disease that is associated with severe chronic illness and fatalities. Unlike other human-infective nematodes, S. stercoralis cycles through a single free-living generation and thus serves as a genetically tractable model organism for understanding the mechanisms that enable parasitism. Techniques such as CRISPR/Cas9-mediated mutagenesis and transgenesis are now routinely performed in S. stercoralis by introducing exogenous DNA into free-living adults and then screening their F1 progeny for transgenic or mutant larvae. However, transgenesis in S. stercoralis has been severely hindered by the inability to establish stable transgenic lines that can be propagated for multiple generations through a host; to date, studies of transgenic S. stercoralis have been limited to heterogeneous populations of transgenic F1 larvae. Here, we develop an efficient pipeline for the generation of stable transgenic lines in S. stercoralis. We also show that this approach can be used to efficiently generate stable transgenic lines in the rat-infective nematode Strongyloides ratti. The ability to generate stable transgenic lines circumvents the limitations of working with heterogeneous F1 populations, such as variable transgene expression and the inability to generate transgenics of all life stages. Our transgenesis approach will enable novel lines of inquiry into parasite biology, such as transgene-based comparisons between free-living and parasitic generations.
Collapse
Affiliation(s)
- Ruhi Patel
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Astra S Bryant
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | - Michelle L Castelletto
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Breanna Walsh
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Interdepartmental PhD Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Medical Scientist Training Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Damia Akimori
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Interdepartmental PhD Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Elissa A Hallem
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
43
|
Kohlbrenner T, Berger S, Laranjeira AC, Aegerter-Wilmsen T, Comi LF, deMello A, Hajnal A. Actomyosin-mediated apical constriction promotes physiological germ cell death in C. elegans. PLoS Biol 2024; 22:e3002775. [PMID: 39178318 PMCID: PMC11376560 DOI: 10.1371/journal.pbio.3002775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/05/2024] [Accepted: 07/30/2024] [Indexed: 08/25/2024] Open
Abstract
Germ cell apoptosis in Caenorhabditis elegans hermaphrodites is a physiological process eliminating around 60% of all cells in meiotic prophase to maintain tissue homeostasis. In contrast to programmed cell death in the C. elegans soma, the selection of germ cells undergoing apoptosis is stochastic. By live-tracking individual germ cells at the pachytene stage, we found that germ cells smaller than their neighbors are selectively eliminated through apoptosis before differentiating into oocytes. Thus, cell size is a strong predictor of physiological germ cell death. The RAS/MAPK and ECT/RHO/ROCK pathways together regulate germ cell size by controlling actomyosin constriction at the apical rachis bridges, which are cellular openings connecting the syncytial germ cells to a shared cytoplasmic core. Enhancing apical constriction reduces germ cell size and increases the rate of cell death while inhibiting the actomyosin network in the germ cells prevents their death. We propose that actomyosin contractility at the rachis bridges of the syncytial germ cells amplifies intrinsic disparities in cell size. Through this mechanism, the animals can adjust the balance between physiological germ cell death and oocyte differentiation.
Collapse
Affiliation(s)
- Tea Kohlbrenner
- Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
- Molecular Life Science PhD Program, University of Zürich and ETH Zürich, Zürich, Switzerland
| | - Simon Berger
- Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
- Institute for Chemical and Bioengineering, ETH Zürich, Zürich, Switzerland
| | - Ana Cristina Laranjeira
- Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
- Molecular Life Science PhD Program, University of Zürich and ETH Zürich, Zürich, Switzerland
| | | | - Laura Filomena Comi
- Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
- Molecular Life Science PhD Program, University of Zürich and ETH Zürich, Zürich, Switzerland
| | - Andrew deMello
- Institute for Chemical and Bioengineering, ETH Zürich, Zürich, Switzerland
| | - Alex Hajnal
- Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
| |
Collapse
|
44
|
Murari E, Meadows D, Cuda N, Mangone M. A comprehensive analysis of 3'UTRs in Caenorhabditis elegans. Nucleic Acids Res 2024; 52:7523-7538. [PMID: 38917330 PMCID: PMC11260456 DOI: 10.1093/nar/gkae543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/29/2024] [Accepted: 06/11/2024] [Indexed: 06/27/2024] Open
Abstract
3'Untranslated regions (3'UTRs) are essential portions of genes containing elements necessary for pre-mRNA 3'end processing and are involved in post-transcriptional gene regulation. Despite their importance, they remain poorly characterized in eukaryotes. Here, we have used a multi-pronged approach to extract and curate 3'UTR data from 11533 publicly available datasets, corresponding to the entire collection of Caenorhabditis elegans transcriptomes stored in the NCBI repository from 2009 to 2023. We have also performed high throughput cloning pipelines to identify and validate rare 3'UTR isoforms and incorporated and manually curated 3'UTR isoforms from previously published datasets. This updated C. elegans 3'UTRome (v3) is the most comprehensive resource in any metazoan to date, covering 97.4% of the 20362 experimentally validated protein-coding genes with refined and updated 3'UTR boundaries for 23489 3'UTR isoforms. We also used this novel dataset to identify and characterize sequence elements involved in pre-mRNA 3'end processing and update miRNA target predictions. This resource provides important insights into the 3'UTR formation, function, and regulation in eukaryotes.
Collapse
Affiliation(s)
- Emma Murari
- The Biodesign Institute at Arizona State University, 1001 S McAllister Ave, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ, USA
| | - Dalton Meadows
- The Biodesign Institute at Arizona State University, 1001 S McAllister Ave, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ, USA
| | - Nicholas Cuda
- The Biodesign Institute at Arizona State University, 1001 S McAllister Ave, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ, USA
| | - Marco Mangone
- The Biodesign Institute at Arizona State University, 1001 S McAllister Ave, Tempe, AZ, USA
| |
Collapse
|
45
|
Valera-Alberni M, Yao P, Romero-Sanz S, Lanjuin A, Mair WB. Novel Imaging Tools to Study Mitochondrial Dynamics in Caenorhabditis elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.603730. [PMID: 39071403 PMCID: PMC11275731 DOI: 10.1101/2024.07.16.603730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Mitochondria exhibit a close interplay between their structure and function. Understanding this intricate relationship requires advanced imaging techniques that can capture the dynamic nature of mitochondria and their impact on cellular processes. However, much of the work on mitochondrial dynamics has been done in single celled organisms or in vitro cell culture. Here, we introduce novel genetic tools for live imaging of mitochondrial networks in the nematode Caenorhabditis elegans , addressing a pressing need for advanced techniques in studying organelle dynamics within live intact multicellular organisms. Through a comprehensive analysis, we directly compare our tools with existing methods, demonstrating their advantages for visualizing mitochondrial morphology and contrasting their impact on organismal physiology. We reveal limitations of conventional techniques, while showcasing the utility and versatility of our approaches, including endogenous CRISPR tags and ectopic labeling. By providing a guide for selecting the most suitable tools based on experimental goals, our work advances mitochondrial research in C. elegans and enhances the strategic integration of diverse imaging modalities for a holistic understanding of organelle dynamics in living organisms.
Collapse
|
46
|
Yang H, Lee D, Kim H, Cook DE, Paik YK, Andersen EC, Lee J. Glial expression of a steroidogenic enzyme underlies natural variation in hitchhiking behavior. Proc Natl Acad Sci U S A 2024; 121:e2320796121. [PMID: 38959036 PMCID: PMC11252821 DOI: 10.1073/pnas.2320796121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/10/2024] [Indexed: 07/04/2024] Open
Abstract
Phoresy is an interspecies interaction that facilitates spatial dispersal by attaching to a more mobile species. Hitchhiking species have evolved specific traits for physical contact and successful phoresy, but the regulatory mechanisms involved in such traits and their evolution are largely unexplored. The nematode Caenorhabditis elegans displays a hitchhiking behavior known as nictation during its stress-induced developmental stage. Dauer-specific nictation behavior has an important role in natural C. elegans populations, which experience boom-and-bust population dynamics. In this study, we investigated the nictation behavior of 137 wild C. elegans strains sampled throughout the world. We identified species-wide natural variation in nictation and performed a genome-wide association mapping. We show that the variants in the promoter of nta-1, encoding a putative steroidogenic enzyme, underlie differences in nictation. This difference is due to the changes in nta-1 expression in glial cells, which implies that glial steroid metabolism regulates phoretic behavior. Population genetic analysis and geographic distribution patterns suggest that balancing selection maintained two nta-1 haplotypes that existed in ancestral C. elegans populations. Our findings contribute to further understanding of the molecular mechanism of species interaction and the maintenance of genetic diversity within natural populations.
Collapse
Affiliation(s)
- Heeseung Yang
- Department of Biological Sciences, Seoul National University, Seoul08826, Republic of Korea
| | - Daehan Lee
- Department of Molecular Biosciences, Northwestern University, Evanston, IL60208
- Department of Biological Sciences, Sungkyunkwan University, Suwon16419, Republic of Korea
| | - Heekyeong Kim
- Yonsei Proteome Research Center, Yonsei University, Seoul03722, Republic of Korea
| | - Daniel E. Cook
- Department of Molecular Biosciences, Northwestern University, Evanston, IL60208
| | - Young-Ki Paik
- Yonsei Proteome Research Center, Yonsei University, Seoul03722, Republic of Korea
| | - Erik C. Andersen
- Department of Molecular Biosciences, Northwestern University, Evanston, IL60208
| | - Junho Lee
- Department of Biological Sciences, Seoul National University, Seoul08826, Republic of Korea
- Research Institute of Basic Sciences, Seoul National University, Seoul08826, Republic of Korea
| |
Collapse
|
47
|
Wang Z, Ke J, Guo Z, Wang Y, Lei K, Wang S, Chen G, Shen Z, Li W, Ou G. Transposase-assisted tagmentation: an economical and scalable strategy for single-worm whole-genome sequencing. G3 (BETHESDA, MD.) 2024; 14:jkae094. [PMID: 38856093 PMCID: PMC11228870 DOI: 10.1093/g3journal/jkae094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/21/2024] [Indexed: 06/11/2024]
Abstract
AlphaMissense identifies 23 million human missense variants as likely pathogenic, but only 0.1% have been clinically classified. To experimentally validate these predictions, chemical mutagenesis presents a rapid, cost-effective method to produce billions of mutations in model organisms. However, the prohibitive costs and limitations in the throughput of whole-genome sequencing (WGS) technologies, crucial for variant identification, constrain its widespread application. Here, we introduce a Tn5 transposase-assisted tagmentation technique for conducting WGS in Caenorhabditis elegans, Escherichia coli, Saccharomyces cerevisiae, and Chlamydomonas reinhardtii. This method, demands merely 20 min of hands-on time for a single-worm or single-cell clones and incurs a cost below 10 US dollars. It effectively pinpoints causal mutations in mutants defective in cilia or neurotransmitter secretion and in mutants synthetically sterile with a variant analogous to the B-Raf Proto-oncogene, Serine/Threonine Kinase (BRAF) V600E mutation. Integrated with chemical mutagenesis, our approach can generate and identify missense variants economically and efficiently, facilitating experimental investigations of missense variants in diverse species.
Collapse
Affiliation(s)
- Zi Wang
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences and MOE Key Laboratory for Protein Science, McGovern Institute for Brain Research, Tsinghua University, Beijing 100190, China
| | - Jingyi Ke
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences and MOE Key Laboratory for Protein Science, McGovern Institute for Brain Research, Tsinghua University, Beijing 100190, China
| | - Zhengyang Guo
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences and MOE Key Laboratory for Protein Science, McGovern Institute for Brain Research, Tsinghua University, Beijing 100190, China
| | - Yang Wang
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences and MOE Key Laboratory for Protein Science, McGovern Institute for Brain Research, Tsinghua University, Beijing 100190, China
| | - Kexin Lei
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences and MOE Key Laboratory for Protein Science, McGovern Institute for Brain Research, Tsinghua University, Beijing 100190, China
| | - Shimin Wang
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences and MOE Key Laboratory for Protein Science, McGovern Institute for Brain Research, Tsinghua University, Beijing 100190, China
| | - Guanghan Chen
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences and MOE Key Laboratory for Protein Science, McGovern Institute for Brain Research, Tsinghua University, Beijing 100190, China
| | - Zijie Shen
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences and MOE Key Laboratory for Protein Science, McGovern Institute for Brain Research, Tsinghua University, Beijing 100190, China
| | - Wei Li
- School of Medicine, Tsinghua University, Beijing 100190, China
| | - Guangshuo Ou
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences and MOE Key Laboratory for Protein Science, McGovern Institute for Brain Research, Tsinghua University, Beijing 100190, China
| |
Collapse
|
48
|
Lyu H, Moya ND, Andersen EC, Chamberlin HM. Gene duplication and evolutionary plasticity of lin-12/Notch gene function in Caenorhabditis. Genetics 2024; 227:iyae064. [PMID: 38809718 PMCID: PMC11492284 DOI: 10.1093/genetics/iyae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/16/2024] [Indexed: 05/31/2024] Open
Abstract
Gene duplication is an important substrate for the evolution of new gene functions, but the impacts of gene duplicates on their own activities and on the developmental networks in which they act are poorly understood. Here, we use a natural experiment of lin-12/Notch gene duplication within the nematode genus Caenorhabditis, combined with characterization of loss- and gain-of-function mutations, to uncover functional distinctions between the duplicate genes in 1 species (Caenorhabditis briggsae) and their single-copy ortholog in Caenorhabditis elegans. First, using improved genomic sequence and gene model characterization, we confirm that the C. briggsae genome includes 2 complete lin-12 genes, whereas most other genes encoding proteins that participate in the LIN-12 signaling pathway retain a one-to-one orthology with C. elegans. We use CRISPR-mediated genome editing to introduce alleles predicted to cause gain-of-function (gf) or loss-of-function (lf) into each C. briggsae gene and find that the gf mutations uncover functional distinctions not apparent from the lf alleles. Specifically, Cbr-lin-12.1(gf), but not Cbr-lin-12.2(gf), causes developmental defects similar to those observed in Cel-lin-12(gf). In contrast to Cel-lin-12(gf), however, the Cbr-lin-12.1(gf) alleles do not cause dominant phenotypes as compared to the wild type, and the mutant phenotype is observed only when 2 gf alleles are present. Our results demonstrate that gene duplicates can exhibit differential capacities to compensate for each other and to interfere with normal development, and uncover coincident gene duplication and evolution of developmental sensitivity to LIN-12/Notch activity.
Collapse
Affiliation(s)
- Haimeng Lyu
- Department of Molecular Genetics, Ohio State University, 484 W 12th Ave, Columbus, OH 43210, USA
| | - Nicolas D Moya
- Department of Biology, Johns Hopkins University, Bascom UTL 383, 3400 North Charles St., Baltimore, MD 21218, USA
| | - Erik C Andersen
- Department of Biology, Johns Hopkins University, Bascom UTL 383, 3400 North Charles St., Baltimore, MD 21218, USA
| | - Helen M Chamberlin
- Department of Molecular Genetics, Ohio State University, 484 W 12th Ave, Columbus, OH 43210, USA
| |
Collapse
|
49
|
Eijlers P, Al-Khafaji M, Soto-Martin E, Fasimoye R, Stead D, Wenzel M, Müller B, Pettitt J. A nematode-specific ribonucleoprotein complex mediates interactions between the major nematode spliced leader snRNP and its target pre-mRNAs. Nucleic Acids Res 2024; 52:7245-7260. [PMID: 38676950 PMCID: PMC11229312 DOI: 10.1093/nar/gkae321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 04/08/2024] [Accepted: 04/12/2024] [Indexed: 04/29/2024] Open
Abstract
Spliced leader trans-splicing of pre-mRNAs is a critical step in the gene expression of many eukaryotes. How the spliced leader RNA and its target transcripts are brought together to form the trans-spliceosome remains an important unanswered question. Using immunoprecipitation followed by protein analysis via mass spectrometry and RIP-Seq, we show that the nematode-specific proteins, SNA-3 and SUT-1, form a complex with a set of enigmatic non-coding RNAs, the SmY RNAs. Our work redefines the SmY snRNP and shows for the first time that it is essential for nematode viability and is involved in spliced leader trans-splicing. SNA-3 and SUT-1 are associated with the 5' ends of most, if not all, nascent capped RNA polymerase II transcripts, and they also interact with components of the major nematode spliced leader (SL1) snRNP. We show that depletion of SNA-3 impairs the co-immunoprecipitation between one of the SL1 snRNP components, SNA-2, and several core spliceosomal proteins. We thus propose that the SmY snRNP recruits the SL1 snRNP to the 5' ends of nascent pre-mRNAs, an instrumental step in the assembly of the trans-spliceosome.
Collapse
Affiliation(s)
- Peter Eijlers
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD Scotland, UK
| | - Mohammed Al-Khafaji
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD Scotland, UK
| | - Eva Soto-Martin
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD Scotland, UK
| | - Rotimi Fasimoye
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD Scotland, UK
| | - David Stead
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Rowett Institute, Foresterhill, Aberdeen AB25 2ZD Scotland, UK
| | - Marius Wenzel
- School of Biological Sciences, University of Aberdeen, Aberdeen AB24 2TZ Scotland, UK
| | - Berndt Müller
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD Scotland, UK
| | - Jonathan Pettitt
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD Scotland, UK
| |
Collapse
|
50
|
Martin CG, Bent JS, Hill T, Topalidou I, Singhvi A. Epithelial UNC-23 limits mechanical stress to maintain glia-neuron architecture in C. elegans. Dev Cell 2024; 59:1668-1688.e7. [PMID: 38670103 PMCID: PMC11233253 DOI: 10.1016/j.devcel.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/23/2023] [Accepted: 04/03/2024] [Indexed: 04/28/2024]
Abstract
For an organ to maintain correct architecture and function, its diverse cellular components must coordinate their size and shape. Although cell-intrinsic mechanisms driving homotypic cell-cell coordination are known, it is unclear how cell shape is regulated across heterotypic cells. We find that epithelial cells maintain the shape of neighboring sense-organ glia-neuron units in adult Caenorhabditis elegans (C. elegans). Hsp co-chaperone UNC-23/BAG2 prevents epithelial cell shape from deforming, and its loss causes head epithelia to stretch aberrantly during animal movement. In the sense-organ glia, amphid sheath (AMsh), this causes progressive fibroblast growth factor receptor (FGFR)-dependent disruption of the glial apical cytoskeleton. Resultant glial cell shape alteration causes concomitant shape change in glia-associated neuron endings. Epithelial UNC-23 maintenance of glia-neuron shape is specific both spatially, within a defined anatomical zone, and temporally, in a developmentally critical period. As all molecular components uncovered are broadly conserved across central and peripheral nervous systems, we posit that epithelia may similarly regulate glia-neuron architecture cross-species.
Collapse
Affiliation(s)
- Cecilia G Martin
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - James S Bent
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Tyler Hill
- Department of Biology, Brandeis University, Waltham, MA 02454, USA
| | - Irini Topalidou
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Aakanksha Singhvi
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Department of Biological Structure, University of Washington School of Medicine, Seattle, WA 98195, USA.
| |
Collapse
|